WO2024162262A1 - Method for selectively inducing antigen-specific inductive regulatory t cells - Google Patents
Method for selectively inducing antigen-specific inductive regulatory t cells Download PDFInfo
- Publication number
- WO2024162262A1 WO2024162262A1 PCT/JP2024/002648 JP2024002648W WO2024162262A1 WO 2024162262 A1 WO2024162262 A1 WO 2024162262A1 JP 2024002648 W JP2024002648 W JP 2024002648W WO 2024162262 A1 WO2024162262 A1 WO 2024162262A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- antigen
- subject
- antibody
- derived
- Prior art date
Links
- 239000000427 antigen Substances 0.000 title claims abstract description 262
- 108091007433 antigens Proteins 0.000 title claims abstract description 262
- 102000036639 antigens Human genes 0.000 title claims abstract description 262
- 238000000034 method Methods 0.000 title claims abstract description 103
- 230000001939 inductive effect Effects 0.000 title claims abstract description 27
- 210000003289 regulatory T cell Anatomy 0.000 title abstract description 44
- 210000004027 cell Anatomy 0.000 claims abstract description 271
- 239000000126 substance Substances 0.000 claims abstract description 28
- 238000002156 mixing Methods 0.000 claims abstract description 22
- 210000001744 T-lymphocyte Anatomy 0.000 claims description 120
- 108010002350 Interleukin-2 Proteins 0.000 claims description 119
- 102000000588 Interleukin-2 Human genes 0.000 claims description 119
- 230000001105 regulatory effect Effects 0.000 claims description 67
- 239000000203 mixture Substances 0.000 claims description 55
- 238000004519 manufacturing process Methods 0.000 claims description 48
- 230000002401 inhibitory effect Effects 0.000 claims description 47
- 239000003112 inhibitor Substances 0.000 claims description 40
- 230000028993 immune response Effects 0.000 claims description 31
- 230000003993 interaction Effects 0.000 claims description 31
- 108020001507 fusion proteins Proteins 0.000 claims description 28
- 102000037865 fusion proteins Human genes 0.000 claims description 28
- 210000001519 tissue Anatomy 0.000 claims description 28
- 101000914484 Homo sapiens T-lymphocyte activation antigen CD80 Proteins 0.000 claims description 26
- 102100027222 T-lymphocyte activation antigen CD80 Human genes 0.000 claims description 26
- 108010038453 Interleukin-2 Receptors Proteins 0.000 claims description 22
- 102000010789 Interleukin-2 Receptors Human genes 0.000 claims description 22
- 206010020751 Hypersensitivity Diseases 0.000 claims description 21
- 230000007815 allergy Effects 0.000 claims description 21
- 210000000056 organ Anatomy 0.000 claims description 21
- 101000914514 Homo sapiens T-cell-specific surface glycoprotein CD28 Proteins 0.000 claims description 19
- 102100027213 T-cell-specific surface glycoprotein CD28 Human genes 0.000 claims description 19
- 239000012634 fragment Substances 0.000 claims description 17
- 208000023275 Autoimmune disease Diseases 0.000 claims description 16
- 201000010099 disease Diseases 0.000 claims description 16
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 16
- 208000026935 allergic disease Diseases 0.000 claims description 13
- 230000004073 interleukin-2 production Effects 0.000 claims description 12
- -1 small molecule compound Chemical class 0.000 claims description 12
- 238000002054 transplantation Methods 0.000 claims description 12
- 208000009329 Graft vs Host Disease Diseases 0.000 claims description 11
- 208000024908 graft versus host disease Diseases 0.000 claims description 11
- 101001057504 Homo sapiens Interferon-stimulated gene 20 kDa protein Proteins 0.000 claims description 8
- 101001055144 Homo sapiens Interleukin-2 receptor subunit alpha Proteins 0.000 claims description 8
- 102100026878 Interleukin-2 receptor subunit alpha Human genes 0.000 claims description 8
- 230000009471 action Effects 0.000 claims description 6
- 230000001404 mediated effect Effects 0.000 claims description 6
- 206010011968 Decreased immune responsiveness Diseases 0.000 description 28
- 210000004989 spleen cell Anatomy 0.000 description 27
- 230000006870 function Effects 0.000 description 26
- 210000004698 lymphocyte Anatomy 0.000 description 26
- 239000006285 cell suspension Substances 0.000 description 25
- 241000699666 Mus <mouse, genus> Species 0.000 description 21
- 230000035755 proliferation Effects 0.000 description 19
- 239000006228 supernatant Substances 0.000 description 17
- 230000006058 immune tolerance Effects 0.000 description 15
- 210000003819 peripheral blood mononuclear cell Anatomy 0.000 description 15
- 239000001963 growth medium Substances 0.000 description 14
- 108090000623 proteins and genes Proteins 0.000 description 14
- 239000008188 pellet Substances 0.000 description 13
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 11
- 239000011780 sodium chloride Substances 0.000 description 11
- 102100037850 Interferon gamma Human genes 0.000 description 10
- 108010074328 Interferon-gamma Proteins 0.000 description 10
- 239000003153 chemical reaction reagent Substances 0.000 description 10
- 238000007710 freezing Methods 0.000 description 10
- 230000008014 freezing Effects 0.000 description 10
- 102000004169 proteins and genes Human genes 0.000 description 10
- 241000699670 Mus sp. Species 0.000 description 9
- 230000000694 effects Effects 0.000 description 9
- 239000002158 endotoxin Substances 0.000 description 9
- 238000011282 treatment Methods 0.000 description 9
- 239000012228 culture supernatant Substances 0.000 description 8
- 239000003814 drug Substances 0.000 description 8
- 238000007799 mixed lymphocyte reaction assay Methods 0.000 description 8
- 238000002617 apheresis Methods 0.000 description 7
- 239000000243 solution Substances 0.000 description 7
- 230000000638 stimulation Effects 0.000 description 7
- 238000010257 thawing Methods 0.000 description 7
- 108010088751 Albumins Proteins 0.000 description 6
- 102000009027 Albumins Human genes 0.000 description 6
- 229960005347 belatacept Drugs 0.000 description 6
- 238000012258 culturing Methods 0.000 description 6
- 238000002347 injection Methods 0.000 description 6
- 239000007924 injection Substances 0.000 description 6
- 239000002609 medium Substances 0.000 description 6
- 108020004707 nucleic acids Proteins 0.000 description 6
- 150000007523 nucleic acids Chemical class 0.000 description 6
- 102000039446 nucleic acids Human genes 0.000 description 6
- 238000012545 processing Methods 0.000 description 6
- 230000001629 suppression Effects 0.000 description 6
- 238000012360 testing method Methods 0.000 description 6
- 238000002965 ELISA Methods 0.000 description 5
- 108060003951 Immunoglobulin Proteins 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 102000018358 immunoglobulin Human genes 0.000 description 5
- 239000007788 liquid Substances 0.000 description 5
- 230000003472 neutralizing effect Effects 0.000 description 5
- 238000002360 preparation method Methods 0.000 description 5
- 150000003384 small molecules Chemical class 0.000 description 5
- 238000012546 transfer Methods 0.000 description 5
- 238000011740 C57BL/6 mouse Methods 0.000 description 4
- 102100032912 CD44 antigen Human genes 0.000 description 4
- 101000868273 Homo sapiens CD44 antigen Proteins 0.000 description 4
- 239000011324 bead Substances 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 238000011156 evaluation Methods 0.000 description 4
- 230000005764 inhibitory process Effects 0.000 description 4
- 150000002632 lipids Chemical class 0.000 description 4
- 239000000463 material Substances 0.000 description 4
- 238000012986 modification Methods 0.000 description 4
- 230000004048 modification Effects 0.000 description 4
- 210000005087 mononuclear cell Anatomy 0.000 description 4
- 239000002504 physiological saline solution Substances 0.000 description 4
- 230000002265 prevention Effects 0.000 description 4
- 239000000047 product Substances 0.000 description 4
- 230000005855 radiation Effects 0.000 description 4
- 238000002560 therapeutic procedure Methods 0.000 description 4
- 102000008203 CTLA-4 Antigen Human genes 0.000 description 3
- 108010021064 CTLA-4 Antigen Proteins 0.000 description 3
- 229940045513 CTLA4 antagonist Drugs 0.000 description 3
- 241000282472 Canis lupus familiaris Species 0.000 description 3
- 241000282326 Felis catus Species 0.000 description 3
- 102100027581 Forkhead box protein P3 Human genes 0.000 description 3
- 101000861452 Homo sapiens Forkhead box protein P3 Proteins 0.000 description 3
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 3
- 229960003697 abatacept Drugs 0.000 description 3
- 239000002671 adjuvant Substances 0.000 description 3
- 210000000612 antigen-presenting cell Anatomy 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 238000004113 cell culture Methods 0.000 description 3
- 239000002299 complementary DNA Substances 0.000 description 3
- 238000012790 confirmation Methods 0.000 description 3
- 230000000139 costimulatory effect Effects 0.000 description 3
- 210000004443 dendritic cell Anatomy 0.000 description 3
- 238000000432 density-gradient centrifugation Methods 0.000 description 3
- 230000001419 dependent effect Effects 0.000 description 3
- 239000012997 ficoll-paque Substances 0.000 description 3
- 230000014509 gene expression Effects 0.000 description 3
- 210000004408 hybridoma Anatomy 0.000 description 3
- 210000000987 immune system Anatomy 0.000 description 3
- 230000006698 induction Effects 0.000 description 3
- 230000000977 initiatory effect Effects 0.000 description 3
- 210000004185 liver Anatomy 0.000 description 3
- 229910052751 metal Inorganic materials 0.000 description 3
- 239000002184 metal Substances 0.000 description 3
- 150000002739 metals Chemical class 0.000 description 3
- 210000005259 peripheral blood Anatomy 0.000 description 3
- 239000011886 peripheral blood Substances 0.000 description 3
- 230000002829 reductive effect Effects 0.000 description 3
- 238000009517 secondary packaging Methods 0.000 description 3
- 230000019491 signal transduction Effects 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- QCVGEOXPDFCNHA-UHFFFAOYSA-N 5,5-dimethyl-2,4-dioxo-1,3-oxazolidine-3-carboxamide Chemical compound CC1(C)OC(=O)N(C(N)=O)C1=O QCVGEOXPDFCNHA-UHFFFAOYSA-N 0.000 description 2
- 235000009434 Actinidia chinensis Nutrition 0.000 description 2
- 244000298697 Actinidia deliciosa Species 0.000 description 2
- 235000009436 Actinidia deliciosa Nutrition 0.000 description 2
- 244000144730 Amygdalus persica Species 0.000 description 2
- 235000017060 Arachis glabrata Nutrition 0.000 description 2
- 244000105624 Arachis hypogaea Species 0.000 description 2
- 235000010777 Arachis hypogaea Nutrition 0.000 description 2
- 235000018262 Arachis monticola Nutrition 0.000 description 2
- 241000972773 Aulopiformes Species 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 241000282693 Cercopithecidae Species 0.000 description 2
- 241000238424 Crustacea Species 0.000 description 2
- 108020004414 DNA Proteins 0.000 description 2
- 102000002322 Egg Proteins Human genes 0.000 description 2
- 108010000912 Egg Proteins Proteins 0.000 description 2
- 240000008620 Fagopyrum esculentum Species 0.000 description 2
- 235000009419 Fagopyrum esculentum Nutrition 0.000 description 2
- 241000287828 Gallus gallus Species 0.000 description 2
- 244000068988 Glycine max Species 0.000 description 2
- 235000010469 Glycine max Nutrition 0.000 description 2
- 102000001398 Granzyme Human genes 0.000 description 2
- 108060005986 Granzyme Proteins 0.000 description 2
- 241000238631 Hexapoda Species 0.000 description 2
- 235000007688 Lycopersicon esculentum Nutrition 0.000 description 2
- 240000008790 Musa x paradisiaca Species 0.000 description 2
- 235000018290 Musa x paradisiaca Nutrition 0.000 description 2
- 241000204031 Mycoplasma Species 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 240000007594 Oryza sativa Species 0.000 description 2
- 235000007164 Oryza sativa Nutrition 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- 235000006040 Prunus persica var persica Nutrition 0.000 description 2
- 201000004681 Psoriasis Diseases 0.000 description 2
- 239000012979 RPMI medium Substances 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 206010039710 Scleroderma Diseases 0.000 description 2
- 241000269821 Scombridae Species 0.000 description 2
- 244000000231 Sesamum indicum Species 0.000 description 2
- 235000003434 Sesamum indicum Nutrition 0.000 description 2
- 240000003768 Solanum lycopersicum Species 0.000 description 2
- 230000006044 T cell activation Effects 0.000 description 2
- 235000021307 Triticum Nutrition 0.000 description 2
- 244000098338 Triticum aestivum Species 0.000 description 2
- 206010046851 Uveitis Diseases 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 239000013566 allergen Substances 0.000 description 2
- 229940124599 anti-inflammatory drug Drugs 0.000 description 2
- 230000001580 bacterial effect Effects 0.000 description 2
- 235000015278 beef Nutrition 0.000 description 2
- 230000033228 biological regulation Effects 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 229940046731 calcineurin inhibitors Drugs 0.000 description 2
- 210000004413 cardiac myocyte Anatomy 0.000 description 2
- 210000003321 cartilage cell Anatomy 0.000 description 2
- 230000004663 cell proliferation Effects 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 238000010586 diagram Methods 0.000 description 2
- 238000007599 discharging Methods 0.000 description 2
- 229940079593 drug Drugs 0.000 description 2
- 239000000428 dust Substances 0.000 description 2
- 235000014103 egg white Nutrition 0.000 description 2
- 210000000969 egg white Anatomy 0.000 description 2
- 238000005516 engineering process Methods 0.000 description 2
- 239000013604 expression vector Substances 0.000 description 2
- 238000011049 filling Methods 0.000 description 2
- 230000002538 fungal effect Effects 0.000 description 2
- 239000003102 growth factor Substances 0.000 description 2
- 210000005003 heart tissue Anatomy 0.000 description 2
- 230000002163 immunogen Effects 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 238000002955 isolation Methods 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 239000004816 latex Substances 0.000 description 2
- 229920000126 latex Polymers 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 235000020640 mackerel Nutrition 0.000 description 2
- 210000002540 macrophage Anatomy 0.000 description 2
- 210000004962 mammalian cell Anatomy 0.000 description 2
- 238000005259 measurement Methods 0.000 description 2
- 239000008267 milk Substances 0.000 description 2
- 235000013336 milk Nutrition 0.000 description 2
- 210000004080 milk Anatomy 0.000 description 2
- 230000035772 mutation Effects 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 210000000944 nerve tissue Anatomy 0.000 description 2
- 210000002569 neuron Anatomy 0.000 description 2
- 235000020232 peanut Nutrition 0.000 description 2
- 238000002823 phage display Methods 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 235000015277 pork Nutrition 0.000 description 2
- 238000009117 preventive therapy Methods 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 102000004196 processed proteins & peptides Human genes 0.000 description 2
- 108090000765 processed proteins & peptides Proteins 0.000 description 2
- 230000004853 protein function Effects 0.000 description 2
- 230000004044 response Effects 0.000 description 2
- 235000009566 rice Nutrition 0.000 description 2
- 235000019515 salmon Nutrition 0.000 description 2
- QFJCIRLUMZQUOT-HPLJOQBZSA-N sirolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 QFJCIRLUMZQUOT-HPLJOQBZSA-N 0.000 description 2
- 210000004927 skin cell Anatomy 0.000 description 2
- 230000003637 steroidlike Effects 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 238000011830 transgenic mouse model Methods 0.000 description 2
- 230000014616 translation Effects 0.000 description 2
- 238000005406 washing Methods 0.000 description 2
- ASWBNKHCZGQVJV-UHFFFAOYSA-N (3-hexadecanoyloxy-2-hydroxypropyl) 2-(trimethylazaniumyl)ethyl phosphate Chemical compound CCCCCCCCCCCCCCCC(=O)OCC(O)COP([O-])(=O)OCC[N+](C)(C)C ASWBNKHCZGQVJV-UHFFFAOYSA-N 0.000 description 1
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- VHRSUDSXCMQTMA-PJHHCJLFSA-N 6alpha-methylprednisolone Chemical compound C([C@@]12C)=CC(=O)C=C1[C@@H](C)C[C@@H]1[C@@H]2[C@@H](O)C[C@]2(C)[C@@](O)(C(=O)CO)CC[C@H]21 VHRSUDSXCMQTMA-PJHHCJLFSA-N 0.000 description 1
- 208000026872 Addison Disease Diseases 0.000 description 1
- 208000004300 Atrophic Gastritis Diseases 0.000 description 1
- 206010003827 Autoimmune hepatitis Diseases 0.000 description 1
- 208000023328 Basedow disease Diseases 0.000 description 1
- KUVIULQEHSCUHY-XYWKZLDCSA-N Beclometasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(Cl)[C@@H]1[C@@H]1C[C@H](C)[C@@](C(=O)COC(=O)CC)(OC(=O)CC)[C@@]1(C)C[C@@H]2O KUVIULQEHSCUHY-XYWKZLDCSA-N 0.000 description 1
- VOVIALXJUBGFJZ-KWVAZRHASA-N Budesonide Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@@H]2[C@@H]1[C@@H]1C[C@H]3OC(CCC)O[C@@]3(C(=O)CO)[C@@]1(C)C[C@@H]2O VOVIALXJUBGFJZ-KWVAZRHASA-N 0.000 description 1
- 101100180402 Caenorhabditis elegans jun-1 gene Proteins 0.000 description 1
- 102000004631 Calcineurin Human genes 0.000 description 1
- 108010042955 Calcineurin Proteins 0.000 description 1
- 101710132601 Capsid protein Proteins 0.000 description 1
- 241000251730 Chondrichthyes Species 0.000 description 1
- 101710094648 Coat protein Proteins 0.000 description 1
- 208000015943 Coeliac disease Diseases 0.000 description 1
- 206010009900 Colitis ulcerative Diseases 0.000 description 1
- 208000035473 Communicable disease Diseases 0.000 description 1
- 208000011231 Crohn disease Diseases 0.000 description 1
- PMATZTZNYRCHOR-CGLBZJNRSA-N Cyclosporin A Chemical compound CC[C@@H]1NC(=O)[C@H]([C@H](O)[C@H](C)C\C=C\C)N(C)C(=O)[C@H](C(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](CC(C)C)N(C)C(=O)[C@@H](C)NC(=O)[C@H](C)NC(=O)[C@H](CC(C)C)N(C)C(=O)[C@H](C(C)C)NC(=O)[C@H](CC(C)C)N(C)C(=O)CN(C)C1=O PMATZTZNYRCHOR-CGLBZJNRSA-N 0.000 description 1
- 108010036949 Cyclosporine Proteins 0.000 description 1
- WYQPLTPSGFELIB-JTQPXKBDSA-N Difluprednate Chemical compound C1([C@@H](F)C2)=CC(=O)C=C[C@]1(C)[C@]1(F)[C@@H]2[C@@H]2CC[C@@](C(=O)COC(C)=O)(OC(=O)CCC)[C@@]2(C)C[C@@H]1O WYQPLTPSGFELIB-JTQPXKBDSA-N 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000588724 Escherichia coli Species 0.000 description 1
- HKVAMNSJSFKALM-GKUWKFKPSA-N Everolimus Chemical compound C1C[C@@H](OCCO)[C@H](OC)C[C@@H]1C[C@@H](C)[C@H]1OC(=O)[C@@H]2CCCCN2C(=O)C(=O)[C@](O)(O2)[C@H](C)CC[C@H]2C[C@H](OC)/C(C)=C/C=C/C=C/[C@@H](C)C[C@@H](C)C(=O)[C@H](OC)[C@H](O)/C(C)=C/[C@@H](C)C(=O)C1 HKVAMNSJSFKALM-GKUWKFKPSA-N 0.000 description 1
- 241000724791 Filamentous phage Species 0.000 description 1
- WJOHZNCJWYWUJD-IUGZLZTKSA-N Fluocinonide Chemical compound C1([C@@H](F)C2)=CC(=O)C=C[C@]1(C)[C@]1(F)[C@@H]2[C@@H]2C[C@H]3OC(C)(C)O[C@@]3(C(=O)COC(=O)C)[C@@]2(C)C[C@@H]1O WJOHZNCJWYWUJD-IUGZLZTKSA-N 0.000 description 1
- POPFMWWJOGLOIF-XWCQMRHXSA-N Flurandrenolide Chemical compound C1([C@@H](F)C2)=CC(=O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2C[C@H]3OC(C)(C)O[C@@]3(C(=O)CO)[C@@]2(C)C[C@@H]1O POPFMWWJOGLOIF-XWCQMRHXSA-N 0.000 description 1
- 208000007882 Gastritis Diseases 0.000 description 1
- 208000036495 Gastritis atrophic Diseases 0.000 description 1
- 206010018364 Glomerulonephritis Diseases 0.000 description 1
- 102100021181 Golgi phosphoprotein 3 Human genes 0.000 description 1
- 208000024869 Goodpasture syndrome Diseases 0.000 description 1
- 206010072579 Granulomatosis with polyangiitis Diseases 0.000 description 1
- 208000015023 Graves' disease Diseases 0.000 description 1
- 208000035895 Guillain-Barré syndrome Diseases 0.000 description 1
- MUQNGPZZQDCDFT-JNQJZLCISA-N Halcinonide Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@H]3OC(C)(C)O[C@@]3(C(=O)CCl)[C@@]1(C)C[C@@H]2O MUQNGPZZQDCDFT-JNQJZLCISA-N 0.000 description 1
- 208000035186 Hemolytic Autoimmune Anemia Diseases 0.000 description 1
- 206010019663 Hepatic failure Diseases 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000935587 Homo sapiens Flavin reductase (NADPH) Proteins 0.000 description 1
- 101001002657 Homo sapiens Interleukin-2 Proteins 0.000 description 1
- 241000714260 Human T-lymphotropic virus 1 Species 0.000 description 1
- 241000713772 Human immunodeficiency virus 1 Species 0.000 description 1
- 206010021245 Idiopathic thrombocytopenic purpura Diseases 0.000 description 1
- 108700005091 Immunoglobulin Genes Proteins 0.000 description 1
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 1
- 101710125418 Major capsid protein Proteins 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 206010049567 Miller Fisher syndrome Diseases 0.000 description 1
- 206010028665 Myxoedema Diseases 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 101710141454 Nucleoprotein Proteins 0.000 description 1
- 108091034117 Oligonucleotide Proteins 0.000 description 1
- 102000015636 Oligopeptides Human genes 0.000 description 1
- 108010038807 Oligopeptides Proteins 0.000 description 1
- 206010053159 Organ failure Diseases 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 206010034277 Pemphigoid Diseases 0.000 description 1
- 201000011152 Pemphigus Diseases 0.000 description 1
- 208000031845 Pernicious anaemia Diseases 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 101710083689 Probable capsid protein Proteins 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 206010040047 Sepsis Diseases 0.000 description 1
- 208000021386 Sjogren Syndrome Diseases 0.000 description 1
- 206010042742 Sympathetic ophthalmia Diseases 0.000 description 1
- 108091008874 T cell receptors Proteins 0.000 description 1
- QJJXYPPXXYFBGM-LFZNUXCKSA-N Tacrolimus Chemical compound C1C[C@@H](O)[C@H](OC)C[C@@H]1\C=C(/C)[C@@H]1[C@H](C)[C@@H](O)CC(=O)[C@H](CC=C)/C=C(C)/C[C@H](C)C[C@H](OC)[C@H]([C@H](C[C@H]2C)OC)O[C@@]2(O)C(=O)C(=O)N2CCCC[C@H]2C(=O)O1 QJJXYPPXXYFBGM-LFZNUXCKSA-N 0.000 description 1
- 208000031981 Thrombocytopenic Idiopathic Purpura Diseases 0.000 description 1
- 102000004887 Transforming Growth Factor beta Human genes 0.000 description 1
- 108090001012 Transforming Growth Factor beta Proteins 0.000 description 1
- 206010052779 Transplant rejections Diseases 0.000 description 1
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 1
- 208000025865 Ulcer Diseases 0.000 description 1
- 201000006704 Ulcerative Colitis Diseases 0.000 description 1
- 241000700605 Viruses Species 0.000 description 1
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 1
- 230000003213 activating effect Effects 0.000 description 1
- 208000004631 alopecia areata Diseases 0.000 description 1
- 125000003275 alpha amino acid group Chemical group 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- 229960003099 amcinonide Drugs 0.000 description 1
- ILKJAFIWWBXGDU-MOGDOJJUSA-N amcinonide Chemical compound O([C@@]1([C@H](O2)C[C@@H]3[C@@]1(C[C@H](O)[C@]1(F)[C@@]4(C)C=CC(=O)C=C4CC[C@H]13)C)C(=O)COC(=O)C)C12CCCC1 ILKJAFIWWBXGDU-MOGDOJJUSA-N 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 230000002238 attenuated effect Effects 0.000 description 1
- 230000001363 autoimmune Effects 0.000 description 1
- 201000000448 autoimmune hemolytic anemia Diseases 0.000 description 1
- 201000003710 autoimmune thrombocytopenic purpura Diseases 0.000 description 1
- 208000010928 autoimmune thyroid disease Diseases 0.000 description 1
- 229950000210 beclometasone dipropionate Drugs 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 229960002537 betamethasone Drugs 0.000 description 1
- UREBDLICKHMUKA-DVTGEIKXSA-N betamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-DVTGEIKXSA-N 0.000 description 1
- 210000000601 blood cell Anatomy 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 210000002798 bone marrow cell Anatomy 0.000 description 1
- 229960004436 budesonide Drugs 0.000 description 1
- 210000005252 bulbus oculi Anatomy 0.000 description 1
- 201000011510 cancer Diseases 0.000 description 1
- 230000000747 cardiac effect Effects 0.000 description 1
- 210000000845 cartilage Anatomy 0.000 description 1
- 208000016644 chronic atrophic gastritis Diseases 0.000 description 1
- 208000025302 chronic primary adrenal insufficiency Diseases 0.000 description 1
- 229960001265 ciclosporin Drugs 0.000 description 1
- 229960004703 clobetasol propionate Drugs 0.000 description 1
- CBGUOGMQLZIXBE-XGQKBEPLSA-N clobetasol propionate Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@H](C)[C@@](C(=O)CCl)(OC(=O)CC)[C@@]1(C)C[C@@H]2O CBGUOGMQLZIXBE-XGQKBEPLSA-N 0.000 description 1
- 238000003501 co-culture Methods 0.000 description 1
- 239000002131 composite material Substances 0.000 description 1
- 238000011109 contamination Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 210000004087 cornea Anatomy 0.000 description 1
- 230000004940 costimulation Effects 0.000 description 1
- 108091008034 costimulatory receptors Proteins 0.000 description 1
- 238000005138 cryopreservation Methods 0.000 description 1
- 239000002577 cryoprotective agent Substances 0.000 description 1
- 238000012136 culture method Methods 0.000 description 1
- 210000004748 cultured cell Anatomy 0.000 description 1
- 229930182912 cyclosporin Natural products 0.000 description 1
- 238000011257 definitive treatment Methods 0.000 description 1
- 201000001981 dermatomyositis Diseases 0.000 description 1
- 229960003662 desonide Drugs 0.000 description 1
- WBGKWQHBNHJJPZ-LECWWXJVSA-N desonide Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@@H]2[C@@H]1[C@@H]1C[C@H]3OC(C)(C)O[C@@]3(C(=O)CO)[C@@]1(C)C[C@@H]2O WBGKWQHBNHJJPZ-LECWWXJVSA-N 0.000 description 1
- 229960002593 desoximetasone Drugs 0.000 description 1
- VWVSBHGCDBMOOT-IIEHVVJPSA-N desoximetasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@H](C(=O)CO)[C@@]1(C)C[C@@H]2O VWVSBHGCDBMOOT-IIEHVVJPSA-N 0.000 description 1
- 229960003957 dexamethasone Drugs 0.000 description 1
- UREBDLICKHMUKA-CXSFZGCWSA-N dexamethasone Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@@H](C)[C@@](C(=O)CO)(O)[C@@]1(C)C[C@@H]2O UREBDLICKHMUKA-CXSFZGCWSA-N 0.000 description 1
- 229960004875 difluprednate Drugs 0.000 description 1
- 230000008034 disappearance Effects 0.000 description 1
- 230000003828 downregulation Effects 0.000 description 1
- 238000002651 drug therapy Methods 0.000 description 1
- 229960004199 dutasteride Drugs 0.000 description 1
- JWJOTENAMICLJG-QWBYCMEYSA-N dutasteride Chemical compound O=C([C@H]1CC[C@H]2[C@H]3[C@@H]([C@]4(C=CC(=O)N[C@@H]4CC3)C)CC[C@@]21C)NC1=CC(C(F)(F)F)=CC=C1C(F)(F)F JWJOTENAMICLJG-QWBYCMEYSA-N 0.000 description 1
- 230000005611 electricity Effects 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 210000003238 esophagus Anatomy 0.000 description 1
- 239000012467 final product Substances 0.000 description 1
- SYWHXTATXSMDSB-GSLJADNHSA-N fludrocortisone acetate Chemical compound C1CC2=CC(=O)CC[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1CC[C@@](C(=O)COC(=O)C)(O)[C@@]1(C)C[C@@H]2O SYWHXTATXSMDSB-GSLJADNHSA-N 0.000 description 1
- 229960004511 fludroxycortide Drugs 0.000 description 1
- 229960001347 fluocinolone acetonide Drugs 0.000 description 1
- FEBLZLNTKCEFIT-VSXGLTOVSA-N fluocinolone acetonide Chemical compound C1([C@@H](F)C2)=CC(=O)C=C[C@]1(C)[C@]1(F)[C@@H]2[C@@H]2C[C@H]3OC(C)(C)O[C@@]3(C(=O)CO)[C@@]2(C)C[C@@H]1O FEBLZLNTKCEFIT-VSXGLTOVSA-N 0.000 description 1
- 229960000785 fluocinonide Drugs 0.000 description 1
- 229960003336 fluorocortisol acetate Drugs 0.000 description 1
- 229960001048 fluorometholone Drugs 0.000 description 1
- FAOZLTXFLGPHNG-KNAQIMQKSA-N fluorometholone Chemical compound C([C@@]12C)=CC(=O)C=C1[C@@H](C)C[C@@H]1[C@]2(F)[C@@H](O)C[C@]2(C)[C@@](O)(C(C)=O)CC[C@H]21 FAOZLTXFLGPHNG-KNAQIMQKSA-N 0.000 description 1
- 235000013305 food Nutrition 0.000 description 1
- 238000009472 formulation Methods 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 150000004676 glycans Chemical class 0.000 description 1
- 229960002383 halcinonide Drugs 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 230000002440 hepatic effect Effects 0.000 description 1
- 229940088597 hormone Drugs 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 229940124452 immunizing agent Drugs 0.000 description 1
- 230000001506 immunosuppresive effect Effects 0.000 description 1
- 238000001727 in vivo Methods 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 238000011221 initial treatment Methods 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 238000007689 inspection Methods 0.000 description 1
- 239000013067 intermediate product Substances 0.000 description 1
- 230000004068 intracellular signaling Effects 0.000 description 1
- 238000010253 intravenous injection Methods 0.000 description 1
- 210000002429 large intestine Anatomy 0.000 description 1
- 201000002364 leukopenia Diseases 0.000 description 1
- 231100001022 leukopenia Toxicity 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 231100000835 liver failure Toxicity 0.000 description 1
- 208000007903 liver failure Diseases 0.000 description 1
- 229960003744 loteprednol etabonate Drugs 0.000 description 1
- DMKSVUSAATWOCU-HROMYWEYSA-N loteprednol etabonate Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@@](C(=O)OCCl)(OC(=O)OCC)[C@@]1(C)C[C@@H]2O DMKSVUSAATWOCU-HROMYWEYSA-N 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 108020004999 messenger RNA Proteins 0.000 description 1
- 229960004584 methylprednisolone Drugs 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 229960002744 mometasone furoate Drugs 0.000 description 1
- WOFMFGQZHJDGCX-ZULDAHANSA-N mometasone furoate Chemical compound O([C@]1([C@@]2(C)C[C@H](O)[C@]3(Cl)[C@@]4(C)C=CC(=O)C=C4CC[C@H]3[C@@H]2C[C@H]1C)C(=O)CCl)C(=O)C1=CC=CO1 WOFMFGQZHJDGCX-ZULDAHANSA-N 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- 206010028417 myasthenia gravis Diseases 0.000 description 1
- 208000003786 myxedema Diseases 0.000 description 1
- 201000008383 nephritis Diseases 0.000 description 1
- 210000005036 nerve Anatomy 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 229920001542 oligosaccharide Polymers 0.000 description 1
- 150000002482 oligosaccharides Chemical class 0.000 description 1
- 208000005963 oophoritis Diseases 0.000 description 1
- 229940035567 orencia Drugs 0.000 description 1
- 150000002894 organic compounds Chemical class 0.000 description 1
- 229940043515 other immunoglobulins in atc Drugs 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 201000001976 pemphigus vulgaris Diseases 0.000 description 1
- 210000005105 peripheral blood lymphocyte Anatomy 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 208000005987 polymyositis Diseases 0.000 description 1
- 108091033319 polynucleotide Proteins 0.000 description 1
- 102000040430 polynucleotide Human genes 0.000 description 1
- 239000002157 polynucleotide Substances 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 229920001282 polysaccharide Polymers 0.000 description 1
- 239000005017 polysaccharide Substances 0.000 description 1
- 229960005205 prednisolone Drugs 0.000 description 1
- OIGNJSKKLXVSLS-VWUMJDOOSA-N prednisolone Chemical compound O=C1C=C[C@]2(C)[C@H]3[C@@H](O)C[C@](C)([C@@](CC4)(O)C(=O)CO)[C@@H]4[C@@H]3CCC2=C1 OIGNJSKKLXVSLS-VWUMJDOOSA-N 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- ZAHRKKWIAAJSAO-UHFFFAOYSA-N rapamycin Natural products COCC(O)C(=C/C(C)C(=O)CC(OC(=O)C1CCCCN1C(=O)C(=O)C2(O)OC(CC(OC)C(=CC=CC=CC(C)CC(C)C(=O)C)C)CCC2C)C(C)CC3CCC(O)C(C3)OC)C ZAHRKKWIAAJSAO-UHFFFAOYSA-N 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 201000003068 rheumatic fever Diseases 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 238000005070 sampling Methods 0.000 description 1
- 238000012216 screening Methods 0.000 description 1
- 238000000926 separation method Methods 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 229960002930 sirolimus Drugs 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 210000000813 small intestine Anatomy 0.000 description 1
- 238000003756 stirring Methods 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 239000004094 surface-active agent Substances 0.000 description 1
- 201000000596 systemic lupus erythematosus Diseases 0.000 description 1
- 229960001967 tacrolimus Drugs 0.000 description 1
- QJJXYPPXXYFBGM-SHYZHZOCSA-N tacrolimus Natural products CO[C@H]1C[C@H](CC[C@@H]1O)C=C(C)[C@H]2OC(=O)[C@H]3CCCCN3C(=O)C(=O)[C@@]4(O)O[C@@H]([C@H](C[C@H]4C)OC)[C@@H](C[C@H](C)CC(=C[C@@H](CC=C)C(=O)C[C@H](O)[C@H]2C)C)OC QJJXYPPXXYFBGM-SHYZHZOCSA-N 0.000 description 1
- 230000009258 tissue cross reactivity Effects 0.000 description 1
- 229960002117 triamcinolone acetonide Drugs 0.000 description 1
- YNDXUCZADRHECN-JNQJZLCISA-N triamcinolone acetonide Chemical compound C1CC2=CC(=O)C=C[C@]2(C)[C@]2(F)[C@@H]1[C@@H]1C[C@H]3OC(C)(C)O[C@@]3(C(=O)CO)[C@@]1(C)C[C@@H]2O YNDXUCZADRHECN-JNQJZLCISA-N 0.000 description 1
- 231100000397 ulcer Toxicity 0.000 description 1
- 241000701161 unidentified adenovirus Species 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 239000013598 vector Substances 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/12—Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
- A61K35/14—Blood; Artificial blood
- A61K35/17—Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/06—Immunosuppressants, e.g. drugs for graft rejection
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/08—Antiallergic agents
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/24—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
Definitions
- Liver transplantation has been widely used as a definitive treatment for patients with end-stage liver failure. There are 20,000 cases overseas and more than 500 cases in Japan each year.
- Transplantation is one of the primary treatments of choice for end-stage renal, cardiac, hepatic, and pancreatic organ failure, and despite significant advances in the treatment of transplant rejection in recent years, the majority of transplants are ultimately rejected.
- Current immunosuppressive regimens which rely on continuous drug therapy, predispose organ transplant patients to increased susceptibility to infections and cancer, as even the drugs fail to inhibit responses specifically directed against the transplant.
- One technique for inducing immune tolerance is the induction of antigen-specific immune unresponsiveness (anergy) in T cells.
- One method involves co-culturing regulatory T cells (Treg) with donor cells in the presence of IL-2, and using the donor-specific Tregs that have been divided and proliferated for treatment, while another method involves administering a low dose of IL-2 to proliferate Tregs in vivo and using them for treatment.
- these methods have problems, such as the risk of IL-2 activating other T cells and the proliferation of polyclonal Tregs that are not donor-specific.
- antigen-specific induced suppressor T cells can be selectively induced by contacting cells with a specific factor. Furthermore, the inventors have discovered that regulating the production or function of IL-2 is important in inducing antigen-specific induced suppressor T cells, and have also discovered a new method for inducing antigen-specific induced suppressor T cells by regulating the production or function of IL-2.
- the present invention provides, for example: (Item 1) A method for selectively inducing antigen-specific induced inhibitory T cells from cells derived from a subject, the method comprising a step of mixing a factor that selectively induces antigen-specific induced inhibitory T cells, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a material containing an antigen not derived from the subject. (Item 2) The method according to the above item, wherein the factor that selectively induces antigen-specific induced suppressor T cells is a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2.
- the factor that selectively induces antigen-specific induced suppressor T cells is an inhibitory factor capable of inhibiting the production of IL-2 or an inhibitory factor capable of inhibiting the function of the produced IL-2.
- the factor that selectively induces antigen-specific induced suppressor T cells is an inhibitory factor capable of inhibiting the interaction between IL-2 and IL-2 receptor (IL-2R) and/or an inhibitory factor capable of inhibiting the interaction between CD80 and/or CD86 and CD28.
- IL-2R IL-2 receptor
- the method according to any one of the above items, wherein the factor that selectively induces antigen-specific induced inhibitory T cells is an inhibitor capable of inhibiting the interaction between IL-2 and IL-2R, selected from an anti-IL-2 antibody, an anti-IL-2R antibody, an anti-IL-2R ⁇ (CD25) antibody, an anti-IL-2R ⁇ (CD122) antibody, or an antigen-binding fragment thereof.
- the factor that selectively induces antigen-specific induced inhibitory T cells is an inhibitor capable of inhibiting the interaction between CD80 and/or CD86 and CD28, selected from the group consisting of an anti-CD80 antibody, an anti-CD86 antibody, a bispecific antibody against CD80 and CD86, an anti-CD28 antibody or an antigen-binding fragment thereof, a CTLA4-Ig fusion protein, and a CD28-Ig fusion protein.
- a method for producing induced suppressor T cells from cells derived from a subject comprising the step of mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
- the regulatory factor capable of regulating the production of IL-2 is selected from an inhibitor capable of inhibiting the interaction of CD80 and/or CD86 with CD28, and a small molecule compound that inhibits IL-2 production.
- the regulator capable of regulating the function of the produced IL-2 is selected from an inhibitor capable of inhibiting the interaction of IL-2 with IL-2 receptor (IL-2R) and a small molecule compound that inhibits the action of IL-2.
- the inhibitor capable of inhibiting the interaction of CD80 and/or CD86 with CD28 is selected from the group consisting of an anti-CD80 antibody, an anti-CD86 antibody, a bispecific antibody against CD80 and CD86, an anti-CD28 antibody or an antigen-binding fragment thereof, a CTLA4-Ig fusion protein, and a CD28-Ig fusion protein.
- compositions for selectively inducing antigen-specific induced suppressor T cells from cells derived from a subject comprising a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, characterized in that the composition is contacted with cells derived from the subject and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
- a composition for treating a disease in a subject mediated by an immune response comprising a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, and antigen-specific induced suppressor T cells selectively induced by mixing cells derived from the subject with an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
- a method for treating a disease in a subject mediated by an immune response comprising the steps of mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen, and administering to the subject antigen-specific induced suppressor T cells selectively induced by the mixing.
- An antigen-specific induced suppressor T cell for treating a disease in a subject mediated by an immune response the antigen-specific induced suppressor T cell being selectively induced by mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
- (Item 13C) Use of antigen-specific induced inhibitory T cells in the manufacture of a medicine for treating a disease in a subject mediated by an immune response, wherein the antigen-specific induced inhibitory T cells are selectively induced by mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
- transplant immune rejection allergy, autoimmune disease, graft-versus-host disease, and immune rejection caused by transplantation of iPS cells or ES cells, or cells, tissues, or organs differentiated therefrom.
- the subject is a human.
- the method disclosed herein makes it possible to selectively induce antigen-specific induced suppressor T cells by contacting cells with a specific factor.
- FIG. 1 shows an outline of the experiment in Example 1.
- Figure 2 shows the antigen-specific suppressive ability of whole cells and Tregs by culturing in the presence of anti-CD80/86 antibodies. The left figure shows the results of confirming Treg proliferation, and the right figure shows a graph of IFN- ⁇ production by stimulation with a stimulator.
- FIG. 3 shows an outline of the experiment in Example 1.
- Figure 4 shows the antigen-specific suppressive ability of whole cells and Tregs when IL-2 was further added to culture in the presence of anti-CD80/86 antibodies. The left figure shows the results of confirming Treg proliferation, and the right figure shows a graph of IFN- ⁇ production by stimulation with a stimulator.
- FIG. 5 shows an outline of the experiment in Example 2.
- Figure 6 shows the antigen-specific suppressive ability of whole cells and Tregs by culturing in the presence of anti-IL-2 antibody.
- the left figure shows the results of confirming Treg proliferation, and the right figure shows a graph of IFN- ⁇ production by stimulation with a stimulator.
- FIG. 7 shows a schematic diagram of acquisition of antigen-specific suppressive ability by anti-CD80/86 antibody.
- FIG. 8 shows a schematic diagram of the acquisition of antigen-specific suppressive ability by anti-IL-2 antibodies.
- FIG. 9 shows a graph of IL-2 production by anti-CD80/86 antibodies.
- FIG. 10 shows a table of IL-2 production by anti-CD80/86 antibodies.
- FIG. 11 shows a table of the percentage of CD4 + T cells in cell populations cultured in the presence of anti-CD80/86 and/or anti-IL-2 antibodies.
- Figure 12 shows confirmation of the antigen-specific suppressive ability of whole cells and Tregs by culture in the presence of anti-CD80/86 antibodies and/or anti-IL-2 antibodies.
- FIG. 13 shows a table summarizing the results of FIG.
- immune tolerance refers to a state in which a specific immune response to a specific antigen is not shown or the specific immune response is suppressed.
- Immune tolerance may mean both or either of a state in which immune cells (especially T cells) do not show a specific immune response to a specific antigen or the specific immune response is suppressed, and a state in which a human does not show a specific immune response to a specific antigen or the specific immune response is suppressed.
- Immune tolerance has attracted attention because it makes it possible to treat immune rejection reactions and allergies by inducing immune tolerance.
- PBMCs or T cells in which immune tolerance has been induced
- anergy PBMCs or T cells
- subject includes domestic animals (e.g., cows, sheep, cats, dogs, horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
- domestic animals e.g., cows, sheep, cats, dogs, horses
- primates e.g., humans and non-human primates such as monkeys
- rabbits e.g., mice and rats
- rodents e.g., mice and rats.
- the subject is a human.
- the term "factor” may refer to any substance or other element (e.g., energy such as light, radioactivity, heat, electricity, etc.) as long as it can achieve the intended purpose.
- substances include, but are not limited to, proteins, polypeptides, oligopeptides, peptides, polynucleotides, oligonucleotides, nucleotides, nucleic acids (e.g., DNA such as cDNA and genomic DNA, and RNA such as mRNA), polysaccharides, oligosaccharides, lipids, small organic molecules (e.g., hormones, ligands, signaling substances, other small organic compounds, molecules synthesized by combinatorial chemistry, small molecules that can be used as pharmaceuticals (e.g., small molecule ligands, etc.)), and composite molecules thereof.
- proteins e.g., proteins, polypeptides, oligopeptides, peptides, polynucleotides, oligonucle
- regulatory factor refers to any type of factor, such as a small molecule, protein, nucleic acid, lipid, or sugar, that positively or negatively regulates a given action (e.g., interaction, signal transduction, protein production, protein function, etc.).
- regulation includes modification or non-modification to any level, such as enhancement (increase), as well as reduction (inhibition, suppression), and disappearance, and as used herein, “regulatory factor” includes “inhibitory factor” and "enhancing factor”, etc.
- the proliferation of induced suppressive T cells specific to a particular antigen is promoted by regulating the production of IL-2 or the function of the produced IL-2.
- IL-2 also promotes the proliferation of induced suppressive T cells nonspecific to a particular antigen, it is possible to suppress the proliferation of nonspecific induced suppressive T cells by regulating the production of IL-2 or the function of the produced IL-2. Since complete inhibition of the production and function of IL-2 also suppresses the proliferation of antigen-specific induced suppressive T cells, it is preferable to have a certain amount of IL-2.
- the methods, factors, and compositions disclosed herein can be adjusted to achieve the presence of a predetermined amount of IL-2.
- the term "inhibitory factor” refers to any type of factor, such as a small molecule, protein, nucleic acid, lipid, or sugar, that can inhibit a certain action (e.g., interaction, signal transduction, protein production, protein function, etc.).
- a certain action e.g., interaction, signal transduction, protein production, protein function, etc.
- anergy is induced in T cells by blocking the interaction between CD80 and/or CD86 on the cell surface and CD28 and inhibiting the CD28 costimulatory signal.
- the inhibitor used to block the interaction between CD80 and/or CD86 and CD28 is selected from the group consisting of a small molecule, a protein, a nucleic acid, a lipid, a sugar, and a combination thereof.
- the protein is an antibody or a variant thereof, or a cell surface molecule or a variant thereof.
- the variant of the antibody is an antigen-binding fragment.
- the variant of the cell surface molecule is a fusion protein.
- the inhibitor is selected from the group consisting of an anti-CD80 antibody, an anti-CD86 antibody, a bispecific antibody against CD80 and CD86, an anti-CD28 antibody or an antigen-binding fragment thereof, a CTLA4-Ig fusion protein, and a CD28-Ig fusion protein.
- the CTLA4-Ig fusion protein is abatacept or belatacept. It is also envisioned that an inhibitor that indirectly inhibits the above interaction (e.g., an inhibitor of an upstream or downstream signal transduction) may also be used in combination.
- antibody a molecule or a group thereof that can specifically bind to a particular epitope on an antigen.
- a full-length antibody i.e., an antibody having an Fc portion
- an antibody lacking an Fc portion may be capable of binding to an antigen of interest, and examples of such an antibody include, but are not limited to, a Fab antibody, an F(ab') 2 antibody, an Fab' antibody, an Fv antibody, and an scFv antibody.
- the antibody may be any type of antibody, i.e., an immunoglobulin known in the art.
- the antibody is an antibody of the isotype IgA, IgD, IgE, IgG, or IgM class.
- the antibody described herein comprises one or more alpha, delta, epsilon, gamma, and/or mu heavy chains.
- the antibody described herein comprises one or more kappa or light chains.
- the antibody is an IgG antibody and is one of the four human subclasses: IgG1, IgG2, IgG3, and IgG4.
- Antibodies contemplated for use in the present disclosure also include camelid-derived antibodies (e.g., VHH antibodies), shark-derived antibodies (e.g., single-chain antibodies), peptibodies, nanobodies (single domain antibodies), minibodies, multispecific antibodies (e.g., bispecific antibodies, diabodies, triabodies, tetrabodies, tandem di-scFv, tandem tri-scFv), and the like, which are known in the art. See, e.g., Kortt et al., Biomol Eng. 2001 18:95-108, (2001) and Todorovska et al., J Immunol Methods. 248:47-66, (2001). The antibody also includes modified or unmodified antibodies.
- camelid-derived antibodies e.g., VHH antibodies
- shark-derived antibodies e.g., single-chain antibodies
- peptibodies e.g., single-chain antibodies
- nanobodies single domain antibodies
- minibodies
- the modified antibody may be bound to an antibody with various molecules such as polyethylene glycol.
- the modified antibody can be obtained by chemically modifying the antibody using a known method. For artificially produced antibodies and various methods of modifying/altering antibodies, see also Biochemistry (2016) 88:380-385.
- the term “antibody” in the narrow sense refers to an immunoglobulin or a group thereof that can specifically bind to a specific epitope on an antigen, and a variant thereof is referred to as a "variant of an antibody".
- the term “antibody” in the narrow sense may be a full-length antibody (i.e., an antibody having an Fc portion), and in the present specification, the term “variant of an antibody” may be a variant lacking the Fc portion of the antibody.
- an antibody in the narrow sense may also be referred to as a full-length antibody, and a variant of an antibody may also be referred to as a variant of a full-length antibody.
- a variant lacking an Fc portion may be capable of binding to an antigen of interest, and examples of such variants include, but are not limited to, Fab antibodies, F(ab') 2 antibodies, Fab' antibodies, Fv antibodies, and scFv antibodies.
- variants of an antibody include modified or unmodified antibodies.
- the modified antibody may be bound to an antibody with various molecules such as polyethylene glycol.
- the modified antibody can be obtained by chemically modifying an antibody using a known method.
- a "polyclonal antibody” can be produced, for example, by administering an immunogen containing an antigen of interest to a mammal (e.g., rat, mouse, rabbit, cow, monkey, etc.), bird, etc., to induce production of polyclonal antibodies specific to the antigen.
- the administration of the immunogen may be by injection of one or more immunizing agents and, if desired, an adjuvant.
- Adjuvants may be used to increase the immune response and may include Freund's adjuvant (complete or incomplete), mineral gels (e.g., aluminum hydroxide), or surfactants (e.g., lysolecithin), etc.
- Immunization protocols are known in the art and may be performed by any method that induces an immune response, tailored to the host organism of choice (Protein Experiment Handbook, Yodosha (2003): 86-91.).
- a "monoclonal antibody” includes a population in which the individual antibodies are substantially identical to a single epitope, except for antibodies with a small amount of naturally occurring mutations.
- the individual antibodies in the population may be substantially identical to a single epitope, except for antibodies with a small amount of naturally occurring mutations.
- Monoclonal antibodies are highly specific and differ from conventional polyclonal antibodies, which typically contain different antibodies that are directed to different epitopes and/or different antibodies that are directed to the same epitope. In addition to their specificity, monoclonal antibodies are useful in that they can be synthesized from hybridoma cultures that are uncontaminated by other immunoglobulins.
- monoclonal may indicate the characteristic of being obtained from a substantially homogeneous population of antibodies, but does not imply that the antibody must be produced by any particular method.
- monoclonal antibodies may be produced by methods similar to the hybridoma method described in "Kohler G, Milstein C., Nature. 1975 Aug 7; 256(5517): 495-497.”
- monoclonal antibodies may be produced by methods similar to the recombinant method described in U.S. Pat. No. 4,816,567.
- monoclonal antibodies may be isolated from phage antibody libraries using methods similar to the techniques described in "Clackson et al., Nature.
- monoclonal antibodies may be produced by methods described in "Protein Experiment Handbook, Yodosha (2003): 92-96.”
- a "chimeric antibody” is, for example, a combination of an antibody variable region and an antibody constant region between different organisms, and can be constructed by recombinant gene technology.
- a mouse-human chimeric antibody can be produced, for example, by the method described in "Roguska et al., Proc Natl Acad Sci USA. 1994Feb 1;91(3):969-973.”
- the basic method for producing a mouse-human chimeric antibody is, for example, to link a mouse leader sequence and a variable region sequence present in a cloned cDNA to a sequence encoding a human antibody constant region already present in an expression vector for mammalian cells.
- the mouse leader sequence and the variable region sequence present in the cloned cDNA may be linked to a sequence encoding a human antibody constant region, and then linked to a mammalian cell expression vector.
- the fragment of the human antibody constant region can be any human antibody H chain constant region or human antibody L chain constant region, for example, C ⁇ 1, C ⁇ 2, C ⁇ 3, or C ⁇ 4 for the human H chain, and C ⁇ or C ⁇ for the L chain.
- a "humanized antibody” is an antibody that has, for example, one or more CDRs from a non-human species, a framework region (FR) from a human immunoglobulin, and a constant region from a human immunoglobulin, and that binds to a desired antigen.
- Antibody humanization can be performed using various techniques known in the art (Almagro et al., Front Biosci. 2008 Jan 1;13:1619-1633.). For example, CDR grafting (Ozaki et al., Blood. 1999 Jun 1; 93(11): 3922-3930.), Re-surfacing (Roguska et al., Proc Natl Acad Sci USA.
- FR shuffling (Damschroder et al., Mol Immunol. 2007 Apr; 44(11): 3049-3060. Epub 2007 Jan 22.) can be used.
- amino acid residues in the human FR regions may be replaced with corresponding residues from the CDR donor antibody. This FR substitution can be performed by methods well known in the art (Riechmann et al., Nature. 1988 Mar 24;332(6162):323-327.).
- FR residues important for antigen binding can be identified by modeling the interactions of CDR and FR residues.
- unusual FR residues at specific positions can be identified by sequence comparison.
- a "human antibody” is, for example, an antibody in which the regions constituting the antibody, including the variable and constant regions of the heavy chain and the variable and constant regions of the light chain, are derived from genes encoding human immunoglobulin.
- Major production methods include the transgenic mouse method for producing human antibodies and the phage display method.
- the transgenic mouse method for producing human antibodies if a functional human Ig gene is introduced into a mouse in which endogenous Ig has been knocked out, human antibodies with diverse antigen-binding abilities are produced instead of mouse antibodies. Furthermore, if this mouse is immunized, human monoclonal antibodies can be obtained by the conventional hybridoma method.
- the phage display method is a system in which a foreign gene is expressed as a fusion protein on the N-terminus of the coat protein (g3p, g10p, etc.) of a filamentous phage, typically an E. coli virus such as M13 or T7, so that the phage does not lose its infectivity.
- a filamentous phage typically an E. coli virus such as M13 or T7, so that the phage does not lose its infectivity.
- it can be produced by the method described in "Vaughan et al., Nat Biotechnol. 1996 Mar;14(3):309-314.”
- antigen-binding fragment of an antibody refers to a fragment of any length that maintains the antibody's ability to bind to an antigen.
- antigen-binding fragments include, but are not limited to, Fab, F(ab') 2 , Fab', Fv, and scFv.
- fusion protein refers to a single protein in which two or more different proteins or fragments thereof are covalently linked, or in which the genes of these proteins are recombinantly expressed together.
- cells derived from a subject refers to cells obtained from a subject to which the composition, pharmaceutical, cell preparation, or cells disclosed herein is administered or which is the subject of the method disclosed herein, or cells derived from cells obtained from the subject.
- antigen derived from a subject refers to an antigen produced by the subject itself that elicits an immune response, for example, an antigen produced by the subject itself that causes an autoimmune disease in a subject with an autoimmune disease.
- antigen not derived from a subject refers to a foreign antigen that can elicit an immune response.
- a substance containing an antigen not derived from a subject refers to any substance or collection of substances that contains an antigen not derived from a subject, such as a cell, cell population, tissue, etc. that expresses an antigen not derived from a subject.
- transplant immune rejection refers to a condition in which the immune system of a subject who has received an organ, tissue, or cell transplant attacks and damages or destroys the transplanted organ, tissue, or cell.
- allergens not derived from the subject that cause allergies are also called allergens, and examples of such antigens include, but are not limited to, dust mite antigens, egg white antigens, milk antigens, wheat antigens, peanut antigens, soybean antigens, buckwheat antigens, sesame antigens, rice antigens, crustacean antigens, kiwi antigens, apple antigens, banana antigens, peach antigens, tomato antigens, tuna antigens, salmon antigens, mackerel antigens, beef antigens, chicken antigens, pork antigens, cat dander antigens, insect antigens, pollen antigens, dog dander antigens, fungal antigens, bacterial antigens, latex, haptens, and metals.
- autoimmune disease refers to any disease in which the immune system mounts an unwanted immune response against one's own cells, tissues, or organs.
- autoimmune diseases include rheumatoid arthritis, multiple sclerosis, type 1 diabetes, inflammatory bowel disease (e.g., Crohn's disease or ulcerative colitis), systemic lupus erythematosus, psoriasis, scleroderma, autoimmune thyroid disease, alopecia areata, Graves' disease, Guillain-Barré syndrome, celiac disease, Sjögren's syndrome, rheumatic fever, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, insulitis, oophoritis, and sepsis.
- focal ulcers include, but are not limited to, focal ulcers, uveitis, phacogenic uveitis, myasthenia gravis, primary myxedema, pernicious anemia, autoimmune hemolytic anemia, Addison's disease, scleroderma, Goodpasture's syndrome, nephritis (e.g., glomerulonephritis), psoriasis, pemphigus vulgaris, pemphigoid, sympathetic ophthalmia, idiopathic thrombocytopenic purpura, idiopathic leukopenia, Wegener's granulomatosis, and poly/dermatomyositis.
- focal ulcers include focal ulcers, uveitis, phacogenic uveitis, myasthenia gravis, primary myxedema, pernicious anemia, autoimmune hemolytic anemia, Addison's disease, scleroderma, Goodpasture's
- graft-versus-host disease refers to an immune response in which a transplanted organ, tissue, or cell attacks, damages, or destroys the cells, tissues, or organs of the recipient.
- immune rejection caused by transplantation of iPS cells or ES cells, or cells, tissues, or organs differentiated from these cells refers to an immune rejection caused by an antigen present in iPS cells or ES cells, or an antigen present in cells, tissues, or organs differentiated from iPS cells or ES cells.
- regulatory T cells also referred to as Tregs
- regulatory T cells refer to T cells that control the immune response to an antigen. Regulatory T cells may be Foxp3 positive. Regulatory T cells may be CD4 positive or CD8 positive. Typically, regulatory T cells may be Foxp3 positive, CD25 positive, and CD4 positive.
- suppressor T cell refers to a T cell that suppresses the immune response to an antigen and/or that does not show or has a suppressed immune response to an antigen.
- antigen-specific suppressor T cells refers to T cells that specifically suppress immune responses to a specific antigen and/or T cells that do not show or are suppressed from showing a specific immune response to a specific antigen.
- induced suppressor T cells refers to suppressor T cells induced by stimulation with the methods, compositions, or factors disclosed herein, and refers to a cell population including regulatory T cells (e.g., FOXP3-positive CD4-positive CD25-positive T cells) and suppressor T cells (e.g., CD4-positive anergy T cells or CD8-positive anergy T cells).
- CD8-positive anergy cells and CD4-positive anergy cells contain many CD44-positive cells, and CD8-positive cells and/or CD4-positive cells may be CD44-positive.
- CD45RA/CD45RO can also be used to confirm the generation of anergy cells.
- CD8-positive anergy T cells and/or CD4-positive anergy T cells are CD45RA-negative and CD45RO-positive.
- antigen-specific induced suppressor T cells refers to induced suppressor T cells that specifically suppress immune responses to a specific antigen and/or that do not show or are suppressed to show a specific immune response to a specific antigen.
- selectively inducing antigen-specific induced suppressive T cells does not necessarily require that the induced suppressive T cells be 100% antigen-specific, but refers to inducing an increase in the proportion of antigen-specific induced suppressive T cells when stimulated with the method, composition, or factor disclosed herein (for example, when cells are contacted with a factor that selectively induces antigen-specific induced suppressive T cells) compared to when not stimulated with the method, composition, or factor disclosed herein (for example, when cells are not contacted with a factor that selectively induces antigen-specific induced suppressive T cells).
- factor that selectively induces antigen-specific induced suppressor T cells refers to any factor that induces an increase in the proportion of antigen-specific induced suppressor T cells compared to when the factor is not in contact with cells, and does not necessarily require that the induced suppressor T cells be 100% antigen-specific.
- the inventors have found that in the induction of inducible suppressor T cells, regulating the production of the growth factor IL-2 or the function of the produced IL-2 induces antigen-specific suppressive ability in the inducible suppressor T cells. Furthermore, the inventors have also found a new culture method for inducing antigen specificity by adding a regulatory factor capable of regulating IL-2 production or a regulatory factor capable of regulating the function of the produced IL-2, with the aim of suppressing the antigen-nonspecific division and proliferation of inducible suppressor T cells.
- the present disclosure provides a method for selectively inducing antigen-specific induced suppressor T cells from cells derived from a subject, the method comprising the step of mixing a factor that selectively induces antigen-specific induced suppressor T cells, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
- the substance containing the antigen may be a cell, which may be irradiated to prevent proliferation and activation of the cell.
- the present disclosure provides a method for producing induced suppressor T cells from cells derived from a subject, the method comprising the step of mixing an inhibitor capable of inhibiting the production of IL-2 or an inhibitor capable of inhibiting the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
- the present disclosure provides a method for suppressing the proliferation of induced suppressive T cells non-specific to a specific antigen in the production of induced suppressive T cells from cells derived from a subject, the method comprising the step of mixing a factor that selectively induces antigen-specific induced suppressive T cells, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
- the method of the present disclosure includes culturing a mixture containing the above-mentioned factor, cells derived from a subject, and antigens derived from the subject or antigens not derived from the subject or antigens contained therein.
- the induced suppressor T cells of the present disclosure can include CD4 positive anergy T cells, CD8 positive T cells, and regulatory T cells.
- the regulatory T cells exhibit cell surface markers such as Foxp3 + , preferably CD4 + , CD25 + , and Foxp3 + .
- the percentage of CD4 + T cells in the culture at the end of the culture using the disclosed methods, factors, and compositions can be about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, or about 50% or more of the total cells. In a preferred embodiment, the percentage of CD4 + T cells in the culture can be about 35% or more of the total cells.
- the percentage of regulatory T cells (e.g., CD4 + Foxp3 + T cells) in the culture at the end of culture using the disclosed methods, factors, and compositions may be about 5% or more, about 10% or more, about 15% or more, about 20% or more, or about 25% or more of total cells. In a preferred embodiment, the percentage of regulatory T cells in the culture may be about 15% or more of total cells.
- the disclosed methods, factors, and compositions can increase the proportion of CD4 + regulatory T cells (e.g., CD4 + Foxp3 + T cells) among CD4 + T cells.
- the proportion of CD4 + regulatory T cells (e.g., CD4 + Foxp3 + T cells) among CD4 + T cells can be about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 80% or more, about 90% or more, or about 100%.
- the percentage of CD4 + regulatory T cells (e.g., CD4 + Foxp3 + T cells) among CD4 + T cells in cultures at the end of culture using the methods, factors, and compositions of the present disclosure may be about 50% or greater.
- the cells can be peripheral blood mononuclear cells (PBMCs), spleen cells, bone marrow cells, lymph node cells, or any combination thereof.
- PBMCs peripheral blood mononuclear cells
- spleen cells spleen cells
- bone marrow cells spleen cells
- lymph node cells or any combination thereof.
- the methods of the present disclosure are performed without the addition of interleukin-2 (IL-2).
- IL-2 interleukin-2
- the factor that selectively induces antigen-specific induced suppressor T cells may be a regulator capable of regulating the production of IL-2 or a regulator capable of regulating the function of the produced IL-2.
- the regulation may preferably be a negative regulation.
- IL-2 also promotes the proliferation of induced suppressor T cells that are non-specific to a particular antigen, so it is possible to suppress the proliferation of non-specific induced suppressor T cells by regulating the production of IL-2 or regulating the function of the produced IL-2. Since complete inhibition of the production and function of IL-2 would also suppress the proliferation of antigen-specific induced suppressor T cells, it is preferable that a certain amount of IL-2 is present.
- the predetermined amount of IL-2 can be about 100 pg/mL or less, about 90 pg/mL or less, about 80 pg/mL or less, about 70 pg/mL or less, about 60 pg/mL or less, about 50 pg/mL or less, about 40 pg/mL or less, about 30 pg/mL or less, about 20 pg/mL or less, about 19 pg/mL or less, about 18 pg/mL or less, about 17 pg/mL or less, about 16 pg/mL or less, about 15 pg/mL or less, about 14 pg/mL or less, about 13 pg/mL or less, about 12 pg/mL or less, about 11 pg/mL or less, about 10 pg/mL or less, about 9 pg/mL or less, about 8 pg/mL or less, about 7 pg/mL or
- the above-mentioned predetermined amount is the amount of IL-2 at a cell concentration of about 0.5 to 2 ⁇ 10 6 cells/mL, and the amount of IL-2 can be appropriately changed depending on the number of cells being cultured.
- the predetermined amount of IL-2 can also differ depending on the source.
- An appropriate predetermined amount of IL-2 can be appropriately determined based on Examples 3 and 6.
- IL-2 or an anti-IL-2 antibody may be added at the beginning or during the culture to achieve a predetermined amount of IL-2 during the culture.
- the suppressive ability of antigen-specific induced suppressive T cells or cell populations containing the same produced by the method or factor of the present disclosure against an antigen may be about 30% or more (i.e., an immune response of 70% or less relative to the standard), about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more, based on the immune response of T cells or cell populations cultured without using the method or factor of the present disclosure.
- the suppressive ability of antigen-specific induced suppressive T cells or cell populations containing the same produced by the method or factor of the present disclosure against an antigen may be about 50% or more.
- the factor that selectively induces antigen-specific induced suppressor T cells may be a regulator or inhibitor that can regulate or inhibit the production of IL-2, or a regulator or inhibitor that can regulate or inhibit the function of the produced IL-2, or a factor that has both functions.
- the factor that selectively induces antigen-specific induced suppressor T cells can be an inhibitory factor that inhibits the interaction between IL-2 and the IL-2 receptor (IL-2R) and/or an inhibitory factor that can inhibit the interaction between CD80 and/or CD86 and CD28.
- IL-2R IL-2 receptor
- inhibitors capable of inhibiting the interaction between IL-2 and IL-2 receptor include, but are not limited to, anti-IL-2 antibodies, anti-IL-2R antibodies, anti-IL-2R ⁇ (CD25) antibodies, anti-IL-2R ⁇ (CD122) antibodies, or antigen-binding fragments thereof.
- inhibitors that can inhibit IL-2 production include inhibitors that can inhibit the interaction between CD80 and/or CD86 and CD28, or small molecule compounds that inhibit IL-2 production.
- Regulatory or inhibitory factors capable of regulating or inhibiting the function of the produced IL-2 include, but are not limited to, inhibitory factors capable of inhibiting the interaction between IL-2 and the IL-2 receptor (IL-2R), small molecule compounds that inhibit the action of IL-2, etc.
- IL-2R IL-2 receptor
- small molecule compounds that inhibit the action of IL-2, etc.
- Small molecule compounds that inhibit IL-2 production include, but are not limited to, calcineurin inhibitors, steroidal anti-inflammatory drugs, and the like.
- Calcineurin inhibitors include tacrolimus, cyclosporine, and the like. Calcineurin dephosphorylates NFAT (nuclear factor of activated T cells), which allows NFAT to enter the nucleus and bind to interleukin-2 promoters. Blocking this process inhibits IL-2 production.
- Steroidal anti-inflammatory drugs include, but are not limited to, mometasone furoate, clobetasol propionate, loteprednol etabonate, difluprednate, dexamethasone, amcinonide, flurandrenolide, prednisolone, fluocinolone acetonide, desonide, triamcinolone acetonide, budesonide, fludrocortisone acetate, fluocinonide, methylprednisolone, betamethasone, desoximethasone, halcinonide, fluorometholone, beclomethasone dipropionate, and dutasteride.
- Small molecule compounds that inhibit the action of IL-2 include rapamycin (sirolimus) and SDZ RAD. Rapamycin suppresses intracellular signaling and cell proliferation, inhibiting the response of lymphocytes to IL-2 and suppressing the activation of T lymphocytes.
- Inhibitory factors capable of inhibiting the interaction of CD80 and/or CD86 with CD28 include, but are not limited to, for example, anti-CD80 antibodies, anti-CD86 antibodies, bispecific antibodies against CD80 and CD86, anti-CD28 antibodies or antigen-binding fragments thereof, CTLA4-Ig fusion proteins, and CD28-Ig fusion proteins.
- the CTLA4-Ig fusion protein may be abatacept or belatacept.
- CD80 and/or CD86 are expressed by antigen-presenting cells
- CD28 is expressed by T cells
- the inhibitor capable of inhibiting the interaction of CD80 and/or CD86 with CD28 may be an anti-CD80 antibody and/or an anti-CD86 antibody or a CTLA4-Ig fusion protein.
- Inhibitors contemplated for use in the present disclosure include CTLA-4 Ig fusion proteins as described above. CTLA-4 Ig fusion proteins function to compete with CD28, a costimulatory receptor on T cells, for binding to CD80/CD86 on antigen-presenting cells, thereby inhibiting T cell activation.
- the CTLA-4 Ig fusion protein is contemplated to be abatacept (Orencia®), belatacept, or Maxy-4.
- Belatacept contains two amino acid substitutions (L104E and A29Y) that significantly increase the binding avidity for CD80 and CD86 (see Davies JK et al., Cell Transplant. (2012); 21(9): 2047-61; Adams AB et al., J Immunol. (2016) 197(6): 2045-50).
- an inhibitor that is expected to have the same effect as the CTLA4-Ig fusion protein is the CD28-Ig fusion protein (Peach RJ et al., J Exp Med. (1994) 180(6): 2049-2058).
- the inhibitors of the present disclosure may also be used in the form of nucleic acids.
- a nucleic acid encoding a CTLA4-Ig fusion protein can be introduced into a cell via an adenovirus vector or the like and expressed. See, for example, Jin YZ et al., Transplant Proc. (2003); 35(8): 3156-9.
- a composition for selectively inducing antigen-specific inducible suppressor T cells from cells derived from a subject comprising a regulator capable of regulating the production of IL-2 or a regulator capable of regulating the function of the produced IL-2, the composition being contacted with cells derived from the subject and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
- the preliminary confirmation can be performed as follows.
- the various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can appropriately modify them to perform the preliminary confirmation manufacturing.
- Infectious disease screening tests are conducted on donors and patients, and donors are confirmed to be negative for HBs antigen, HCV antibody, HIV-1/2, and HTLV-1 antibody.
- donor lymphocytes can be separated as follows: The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can make appropriate modifications to separate donor lymphocytes.
- - Donor lymphocytes are collected into a collection bag by apheresis, and the collection bag is then irradiated.
- the irradiated peripheral blood mononuclear cells are placed in a centrifuge tube containing an appropriate amount of Ficoll-Paque PREMIUM (GE Healthcare #17-5442-02) or Lymphocyte Separation Solution (Nacalai Tesque #20828) or the like (e.g., 20 mL), and centrifuged at 860 G for 20 minutes at 22°C. Discard the supernatant and transfer the cell suspension containing the lymphocyte layer to another centrifuge tube (for example, two 50 mL centrifuge tubes).
- Ficoll-Paque PREMIUM GE Healthcare #17-5442-02
- Lymphocyte Separation Solution Nacalai Tesque #20828
- saline to the centrifuge tube containing the cell suspension (for example, an appropriate amount until the total volume is 50 mL), and mix well by repeatedly aspirating and dispensing with a syringe (for example, a 50 mL syringe with an 18G needle attached) or a pipette. Centrifuge at 500G for 10 minutes at 22°C (you may set the centrifuge accelerator to fast and the brake to fast). Discard the supernatant, add saline again (for example, an appropriate amount until the total volume is 50 mL), and mix the cell pellet well by repeatedly aspirating and dispensing the cell pellet with a pipette.
- the cryopreservation of donor lymphocytes can be performed as follows.
- the various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to cryopreserve donor lymphocytes.
- a freezing bag for example, Frozebag F-050 25 mL freezing bag, Nipro 89-101
- a syringe e.g., a 30 mL syringe fitted with an 18G needle
- ACD liquid (Terumo TP-A05ACD, for example, 2 mL per 15 mL of cell suspension) to the freezing bag containing the cell suspension, sandwich it between ice packs cooled to 4°C, and chill for about 10 minutes.
- a syringe e.g., a 20 mL syringe with an 18G needle
- CP-1 Kinyokuto Pharmaceutical Co., Ltd. 551-27202-4 Cell Cryoprotectant CP-1
- CP-1 Kinyokuto Pharmaceutical Co., Ltd. 551-27202-4 Cell Cryoprotectant CP-1
- a syringe remove all air from the freezing bag and its ports. Seal the freezing bag using a tube sealer and first chill at 4°C for approximately 5-10 minutes, then store in a -80°C freezer.
- thawing of donor lymphocytes can be performed as follows: The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to thaw donor lymphocytes. Thaw the frozen bag of stored donor cells, for example, in a constant temperature bath at 37° C. The subsequent operations are preferably performed under aseptic conditions.
- a syringe e.g., a 50 mL syringe with an 18G needle attached
- withdraw the cell suspension from the thawed freezing bag and transfer it to a centrifuge tube e.g., 12.5 mL each into two 50 mL centrifuge tubes.
- a centrifuge tube e.g. 12.5 mL each into two 50 mL centrifuge tubes.
- albumin solution Nahon Pharmaceutical Co., Ltd. 123146364 Blood Donation Albumin 5% Intravenous Injection 12.5 g/250 mL
- Intravenous Injection 12.5 g/250 mL
- Centrifuge for example, at 600G for 10 minutes at 22°C (for example, preferably with the centrifuge accelerator set to fast and the brake set to slow). Carefully discard the supernatant, and add an appropriate liquid such as albumin-added saline for washing (for example, made from 25 mL of 5% albumin solution and 19 mL of saline) to the cell pellet to suspend it.
- Centrifuge for example, at 600G for 10 minutes at 22°C (for example, preferably with the centrifuge accelerator set to fast and the brake to slow).
- ALyS505N culture medium for example, 10 mL for a 50 mL centrifuge tube
- -Anti-human CD80 antibody e.g., m2D10.4; Cat. No. 16-0809-85, eBioscience
- anti-human CD86 antibody e.g., IT2.2; Cat. No. 16-0869-85, eBioscience
- a culture bag containing ALyS505N-0 culture medium or a liquid equivalent thereto e.g., Nipro 87598 Nipro Medium ALyS505NB10
- a final concentration of, for example, 10 ⁇ g/mL or an inhibitor such as a CTLA4-Ig fusion protein (e.g., belatacept) is added
- the above cell suspension is added to this culture bag by injection with a syringe (e.g., a 20 mL syringe with an 18G needle).
- the total volume of the liquid in the culture bag is about 840 mL.
- the patient's lymphocytes can be separated as follows.
- the various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to separate the patient's lymphocytes.
- Plasma collected from the patient is inactivated by heating it in a constant temperature bath, for example, at 56°C for 30 minutes. If it is not used immediately, it is stored frozen.
- Peripheral blood collected from a patient is placed in a centrifuge tube containing an appropriate amount of a suitable medium, such as Ficoll-Paque (e.g., 20 mL), and centrifuged, for example, at 860 G for 20 minutes at 22°C (e.g., preferably with the accelerator and brake of the centrifuge set to slow). Discard the supernatant and transfer the cell suspension containing the lymphocyte layer to another centrifuge tube (for example, two 50 mL centrifuge tubes). Add saline to the centrifuge tube containing the cell suspension (for example, an appropriate amount until the total volume is 50 mL), and mix well by repeatedly aspirating and dispensing with a pipette.
- a suitable medium such as Ficoll-Paque (e.g., 20 mL)
- centrifuge at 500 G for 10 minutes at 22° C. the accelerator and brake of the centrifuge may be set to fast and fast, respectively.
- Discard the supernatant add saline again (for example, an appropriate amount until the total volume is 50 mL), and mix the cell pellet well by repeatedly aspirating and dispensing the cell pellet with a pipette.
- centrifuge at 500 G for 5 minutes at 22° C. the accelerator and brake of the centrifuge may be set to fast and fast, respectively).
- the cell pellet is suspended in, for example, ALyS505N-0 culture medium (for example, 10 mL) to prepare a cell suspension (for example, add ALyS505N-0 culture medium until the total volume becomes 20 mL). At this point, about 0.5 mL of the cell suspension is withdrawn, and the cell count, viable cell count, and surface antigen expression are confirmed.
- - Inactivated plasma derived from the patient is added to the culture bag containing the donor cells in the ALyS505N-0 culture medium prepared in "3. Thawing of donor lymphocytes" and inhibitors such as antibodies.
- the patient-derived cell suspension is added to the culture bag by injection using a syringe (e.g., a 20 mL syringe with an 18G needle) and the culture bag is sealed using a tube sealer.
- a syringe e.g., a 20 mL syringe with an 18G needle
- the total volume of the culture bag is about 1000 mL.
- Medium exchange (e.g., after 1 week, preferably under sterile conditions)
- the medium exchange can be performed as follows.
- the various values, reagents, procedures, etc. exemplified below are representative examples, and a person skilled in the art can appropriately modify them to exchange the medium.
- centrifuge tubes e.g., four 225 mL centrifuge tubes. Centrifuge, for example, at 600G for 10 minutes at 22°C (for example, preferably with the centrifuge accelerator set to fast and the brake set to fast).
- the cell pellet is suspended in, for example, ALyS505N-0 culture medium to prepare a cell suspension (for example, add ALyS505N-0 culture medium until the total volume becomes 20 mL). At this point, about 0.3 mL of the cell suspension is removed and the cell count and viable cell count are confirmed.
- the cell suspension is added to a culture bag containing ALyS505N-0 culture medium by injection using a syringe (for example, a 20 mL syringe with an 18G needle).
- a diluted solution of anti-human CD80 antibody e.g., 2D10.4
- an diluted solution of anti-human CD86 antibody e.g., IT2.2
- a final concentration of 10 ⁇ g/mL or an inhibitor such as a CTLA4-Ig fusion protein (e.g., belatacept)
- a syringe e.g., a 20 mL syringe attached with an 18G needle.
- thawing of donor lymphocytes, antigen restimulation, and initiation of secondary culture can be performed as follows.
- the various numerical values, reagents, procedures, etc. exemplified below are representative examples, and a person skilled in the art can appropriately modify the thawing of donor lymphocytes, antigen restimulation, and initiation of secondary culture.
- the frozen bag of stored donor cells and the inactivated plasma from the patient are thawed, for example, in a thermostatic bath at 37° C.
- the subsequent operations are preferably performed under aseptic conditions.
- a syringe e.g., a 50 mL syringe with an 18G needle
- withdraw the donor cell suspension from the thawed cryobag and transfer it to a centrifuge tube e.g., two 50 mL centrifuge tubes.
- 5% albumin solution for example, about 50 mL in total for two 50 mL centrifuge tubes
- centrifuge tube containing the donor cell suspension mix well, and then leave to stand for about 5 minutes. For example, centrifuge at 600 G for 10 minutes at 22° C. (set the centrifuge's accelerator to fast and the brake to slow).
- albumin-containing saline for washing prepared, for example, from 25 mL of 5% albumin solution and 19 mL of saline
- albumin-containing saline for washing prepared, for example, from 25 mL of 5% albumin solution and 19 mL of saline
- centrifuge at 600 G for 10 minutes at 22° C. set the centrifuge's accelerator to fast and the brake to slow.
- ALyS505N-0 culture medium for example, 10 mL for a 50 mL centrifuge tube.
- the thawed inactivated plasma from the patient e.g., 10 mL
- a syringe e.g., a 20 mL syringe with an 18 G needle
- the above-mentioned cell suspension is added by injection with a syringe (e.g., a 20 mL syringe with an 18 G needle) to this culture bag.
- the total volume of the liquid in the culture bag is about 1000 mL.
- Culture in a 37°C incubator for, for example, one week.
- test during the secondary culture can be performed as follows.
- the various values, reagents, procedures, etc. exemplified below are representative examples, and a person skilled in the art can appropriately modify them to perform the test during the secondary culture.
- a small amount of culture medium is withdrawn from the culture bag and tested for mycoplasma contamination, etc.
- Collection and filling of cultured lymphocytes (performed under sterile conditions)
- the collection and filling of cultured lymphocytes can be performed as follows.
- the various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to collect and fill cultured lymphocytes.
- centrifuge tubes for example, four 225 mL centrifuge tubes.
- centrifuge (set the centrifuge accelerator to slow and the brake to slow).
- a centrifuge tube e.g., a 50 mL centrifuge tube.
- Add saline to the centrifuge tube containing the cell suspension for example, an appropriate amount until the total volume is 50 mL
- a syringe for example, a 50 mL syringe with an 18G needle attached.
- centrifuge at 500 G for 10 minutes at 22° C. (the accelerator and brake of the centrifuge may be set to fast and fast, respectively).
- Secondary Packaging can be performed as follows: The various numerical values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can appropriately modify them to perform secondary packaging. Typically, enter and print the subject ID, serial number, and expiration date on a label based on the appropriate standard (typically, NUHCPC-M-12-ATREG) and attach the label to the container. - Issue "Dosage and administration, indications or effects, and precautions for use or handling" based on appropriate standards (typically, NUHCPC-PMF-ATREG14). - Place the test item and "usage, dosage, efficacy or effects, and precautions for use or handling" in a plastic bag with a zipper. - Store in a transport container in the monitoring unit until shipment.
- the cell preparation can be produced as follows: The various numerical values exemplified below are representative examples, and a person skilled in the art can appropriately modify them to produce the cell preparation. 1) Approximately 19 days prior to administration, apheresis will be performed on the donor at a medical institution, and the donor apheresis product will be irradiated with 30 Gy of radiation to eliminate cell proliferation ability, and then shipped to a cell processing facility where cell processing will be performed. 2) After receiving the donor apheresis product, the donor mononuclear cells will be separated and collected by density gradient centrifugation at the cell culture processing facility, then divided into two aliquots, frozen, and stored at -80 ⁇ 10°C.
- apheresis is performed on the recipient at a medical institution, and the recipient apheresis product is shipped to a cell culture processing facility where cell processing will be performed.
- the recipient mononuclear cells are separated and collected by density gradient centrifugation and co-cultured with thawed donor mononuclear cells and inhibitors such as anti-CD80 and anti-CD86 antibodies or CTLA4-Ig fusion protein.
- the medium is replaced about 7 days before administration.
- the intermediate product cultured for 7 days is collected and co-cultured with thawed donor mononuclear cells and inhibitors such as anti-CD80 and anti-CD86 antibodies or CTLA4-Ig fusion protein. 6) On the day of administration, the processed cells are collected by density gradient centrifugation, washed, and filled with physiological saline. 7) Ship it to a medical institution and administer it to the recipient at the medical institution.
- the disclosure provides a composition for treating a disease in a subject mediated by an immune response, the composition comprising an inhibitor that inhibits the interaction of IL-2 with the IL-2 receptor (IL-2R) and/or an inhibitor that can inhibit the interaction of CD80 and/or CD86 with CD28, and antigen-specific suppressor T cells selectively induced by mixing cells derived from the subject with an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
- IL-2R IL-2 receptor
- the disease may be selected from the group consisting of transplant immune rejection, allergy, autoimmune disease, graft-versus-host disease, and immune rejection caused by transplantation of iPS cells or ES cells or cells, tissues, or organs differentiated therefrom.
- the transplant immune rejection is characterized as occurring following transplantation of kidney, liver, heart, skin, lung, pancreas, esophagus, stomach, small intestine, large intestine, nerves, blood, blood cells including immune system cells, bone, cartilage, blood vessels, cornea, eyeball, or bone marrow.
- anergy cells can be induced by mixing the inhibitor, recipient-derived cells, and donor-derived antigens or donor-derived antigen-containing material.
- the donor-derived antigen-containing material can be PBMCs, spleen cells, or cells derived from the organ to be transplanted.
- anergy cells can be induced by mixing the inhibitor, cells derived from the subject, and an antigen not derived from the subject that causes the allergy.
- anergy cells can be induced by mixing the inhibitor, cells derived from the subject, and an antigen derived from the subject that causes the autoimmune disease.
- anergy cells can be induced by mixing the inhibitor, PBMCs or spleen cells of the donor providing the graft, and a recipient-derived antigen or a substance containing the antigen.
- the recipient-derived antigen can be PBMCs, spleen cells, or cells in the vicinity of the site where the organ is transplanted or cells derived therefrom.
- anergy cells can be induced by mixing the above-mentioned inhibitor with cells derived from the subject and cells differentiated from iPS cells or ES cells to be used for transplantation.
- the present disclosure provides a method for treating or preventing allergies and/or autoimmune diseases using the pharmaceutical of the present disclosure, and a pharmaceutical, composition, and cell mixture for use therein.
- allergies and autoimmune diseases macrophages obtained from the peripheral blood of a patient are differentiated into dendritic cells (macrophage-derived dendritic cells) with high antigen-presenting ability by a conventional method, and these cells are made to present an antigen causing an overreaction in allergies or autoimmune diseases after irradiation (gamma rays), and co-cultured with a group of T cells obtained from the peripheral blood of the same patient in the presence of the above factors for 1 to 2 weeks to obtain anergy cells specific to the antigen causing the allergy or autoimmune disease.
- dendritic cells macrophage-derived dendritic cells
- gamma rays gamma rays
- anergy cells By administering these anergy cells to a patient, immune tolerance specific to the antigen causing the allergy or autoimmune disease is induced, and the anergy cells are used for the prevention and treatment of allergies and autoimmune diseases.
- the number of administrations may be multiple times depending on various conditions such as whether it is a preventive therapy or a treatment, and the strength of the symptoms.
- the present disclosure provides a method for treating or preventing graft-versus-host disease using the medicament of the present disclosure, and a medicament, composition, and cell mixture for use therein.
- cells that can cause graft-versus-host disease such as PBMCs or T cells of a donor providing a graft
- the reaction of the graft causing graft-versus-host disease to the host is suppressed (immune tolerance is induced), and graft-versus-host disease is prevented and treated.
- the number of administrations may be multiple, depending on various conditions, such as whether it is a preventive therapy or a treatment, the tissue to be transplanted, its size, and the severity of symptoms.
- the present disclosure provides a method for treating or preventing immune rejection or other side effects caused by transplantation of iPS cells or ES cells and cells, tissues or organs differentiated therefrom in prevention or therapy using iPS cells or ES cells using the pharmaceutical of the present disclosure, and a pharmaceutical, composition, and cell mixture used therein.
- immune rejection caused by transplantation of iPS cells or ES cells and cells, tissues or organs differentiated therefrom is typically exemplified as a treatment target.
- cells or dendritic cells to be used for transplantation that have been differentiated from iPS cells or ES cells are irradiated with radiation (gamma rays), and these cells are co-cultured with the PBMC or T cell group of the patient receiving the transplant in the presence of the above factors for 1-2 weeks to obtain anergy cells specific to the cells differentiated from iPS cells or ES cells.
- radiation gamma rays
- anergy cells specific to the cells differentiated from iPS cells or ES cells By administering these anergy cells to the host, specific immune tolerance is induced to the transplanted cells, tissues, and organs that have been differentiated from iPS cells or ES cells, and rejection reactions to them are prevented and treated.
- the number of administrations may be multiple depending on whether it is a preventive or therapeutic therapy, and on various conditions such as the tissue to be transplanted, its size, and the severity of symptoms.
- the present disclosure provides a method for treating or preventing allergy using the pharmaceutical of the present disclosure, and a pharmaceutical, composition, and cell mixture for use therein.
- the present inventors have demonstrated that a preparation containing cells in which anergy is induced antigen-specifically by the above-mentioned factor can induce immune tolerance to allergy. Therefore, in another aspect, the present disclosure provides a composition for treating or preventing allergy in a subject, the composition comprising cells in which immune tolerance is induced by mixing the above-mentioned factor, cells derived from the subject, and an antigen causing allergy or a substance containing the antigen.
- Allergens that cause allergies include foods, pollen, medicines, and metals, and more specifically include, but are not limited to, dust mite antigens, egg white antigens, milk antigens, wheat antigens, peanut antigens, soybean antigens, buckwheat antigens, sesame antigens, rice antigens, crustacean antigens, kiwi antigens, apple antigens, banana antigens, peach antigens, tomato antigens, tuna antigens, salmon antigens, mackerel antigens, beef antigens, chicken antigens, pork antigens, cat dander antigens, insect antigens, pollen antigens, dog dander antigens, fungal antigens, bacterial antigens, latex, haptens, and metals.
- the present disclosure provides a method for suppressing or preventing immune rejection caused by iPS cells, etc., and provides a medicine, composition, and cell mixture for the suppression or prevention.
- the present inventors have demonstrated that a preparation containing cells in which anergy is induced by the above-mentioned factor can induce immune tolerance to immune rejection caused by iPS cells, etc., or cells, tissues, or organs differentiated therefrom.
- the present disclosure provides a composition for suppressing or preventing immune rejection caused by iPS cells or ES cells, or cells, tissues, or organs differentiated therefrom, in a subject, the composition comprising cells in which immune tolerance has been induced by mixing the above-mentioned factor, cells derived from the subject, and an antigen differentiated from the iPS cells or ES cells, or a substance containing the antigen.
- Cells, tissues, or organs differentiated from iPS cells or ES cells include, but are not limited to, nerve cells or tissue, corneal cells or tissue, cardiac muscle cells or tissue, liver or tissue, cartilage cells or tissue, skin cells or tissue, kidney or tissue, etc.
- cells, tissues, or organs differentiated from iPS cells or ES cells include nerve cells or tissue, cardiac muscle cells or tissue, cartilage cells or tissue, and skin cells or tissue.
- Example 1 Effect of addition of IL-2 on allospecific suppression by Treg
- MLR mixed lymphocyte reaction
- Spleen cells were collected from Balb/c mice and irradiated with 30 Gy of X-rays, after which they were adjusted to a concentration of 4 x 106 cells/mL and mixed with hCD2-KI mouse spleen cells at a 1:1 ratio.
- Anti-CD80 mAb + anti-CD86 mAb (final concentration: 10 ⁇ g/mL each) and, if necessary, recombinant IL-2 (final concentration: 25 U/mL) were added, seeded on a 10 cm dish, and cultured at 37° C. in a 5% CO 2 incubator for 7 days.
- B6 mouse spleen cells, LPS-stimulated Balb/c mouse spleen cells, or LPS-stimulated CBA/Nslc mouse spleen cells, and the cultured whole cells or isolated Tregs from 3. were prepared to the concentrations shown in Table 1 and seeded onto a U-bottom 96-well plate.
- the cells were cultured at 37°C in a 5% CO2 incubator for 3 days.
- Spleen cells were collected from Balb/c mice and irradiated with 30 Gy of X-rays, then adjusted to a concentration of 2 x 106 cells/mL and mixed with hCD2-KI mouse spleen cells at a 1:1 ratio.
- Anti-CD80 mAb + anti-CD86 mAb (final concentration 10 ⁇ g/mL each) or anti-IL-2 mAb (final concentration 10 ⁇ g/mL), and recombinant IL-2 (final concentration 25 U/mL) were added as required, and 200 ⁇ L each was seeded into a U-bottom 96-well plate and cultured at 37°C in a 5% CO2 incubator for 4 days.
- Example 2 Evaluation of allospecific suppressive ability of Treg in MLR with addition of anti-IL-2 antibody
- Example 2 Evaluation of allospecific suppressive ability of Treg in MLR with addition of anti-IL-2 antibody
- Spleen cells were collected from Balb/c mice and irradiated with 30 Gy of X-rays, after which they were adjusted to a concentration of 4 x 106 cells/mL and mixed with hCD2-KI mouse spleen cells at a 1:1 ratio.
- Anti-CD80 mAb + anti-CD86 mAb (final concentration 10 ⁇ g/mL each) or anti-IL-2 mAb (final concentration 10 ⁇ g/mL) was added, seeded on a U-bottom 96-well plate, and cultured at 37° C. in a 5% CO 2 incubator for 7 days.
- B6 mouse spleen cells, LPS-stimulated Balb/c mouse spleen cells, or LPS-stimulated CBA/Nslc mouse spleen cells, and the cultured whole cells or isolated Tregs from 3. were prepared to the concentrations shown in Table 2 and seeded onto a U-bottom 96-well plate.
- the cells were cultured at 37°C in a 5% CO2 incubator for 3 days.
- Example 3 Effect of IL-2 during culture
- the effect of IL-2 on the induction of antigen-specific inducible suppressor T cells using anti-CD80/86 antibody and/or anti-IL-2 antibody was confirmed.
- the method was the same as in Examples 1 and 2.
- the concentration of IL-2 in the culture medium on days 1 to 5 of culture was measured by ELISA.
- (result) 9 and 10 show that when cells were cultured in the presence of anti-CD80/86 antibody or anti-IL-2 antibody, the IL-2 concentration was 20 pg/mL or less on day 5. It is believed that the CD28 costimulatory signal was inhibited by anti-CD80/86 antibody, which reduced T cell activation and suppressed IL-2 production. On the other hand, when cells were cultured without anti-CD80/86 antibody or anti-IL-2 antibody, the IL-2 concentration exceeded 100 pg/mL already on day 2 of culture.
- Example 4 Change in the percentage of CD4 + T cells by anti-CD80/86 antibody or anti-IL-2 antibody
- changes in the proportion of CD4 + T cells due to anti-CD80/86 antibody and/or anti-IL-2 antibody were confirmed.
- the methods were similar to those in Examples 1 and 2.
- Example 5 Change in suppressive ability by anti-CD80/86 antibody or anti-IL-2 antibody
- changes in suppressive ability due to anti-CD80/86 antibody and/or anti-IL-2 antibody were confirmed.
- Tregs When IL-2 was completely blocked using both anti-CD80/86 antibody and anti-IL-2 antibody, isolated Tregs exhibited suppressive ability against Balb stimulation, but whole cells exhibited almost no suppressive ability. When cultured with only anti-CD80/86 antibody, stronger suppressive ability against Balb stimulation was observed. These results indicate that the suppressive ability of suppressive T cells is IL-2 dependent. Isolated Tregs also exhibited slight suppressive ability against CBA, but whole cells exhibited almost no suppressive ability (FIGS. 12 and 13). Therefore, the suppressive ability of whole cells has high antigen specificity and is also highly dependent on IL-2. Antigen non-specific suppression may be due to the influence of the suppressive cytokine TGF- ⁇ , etc.
- Example 6 Evaluation of inhibition of IL-2 production by anti-CD80/86 antibody or anti-IL-2 neutralizing antibody In this example, using human cells, it is confirmed that IL-2 production is suppressed by anti-CD80/86 antibody or anti-IL-2 neutralizing antibody.
- the culture supernatant is collected on days 1, 2, 3, and 5 of culture, and frozen and stored at -20°C until the day of measurement of IL-2 by ELISA. 5. Measure human IL-2 by ELISA.
- the IL-2 concentration in the culture supernatant is predicted to be lower than that in the control group.
- Example 7 Evaluation of the effect of adjusting the IL-2 concentration in the culture medium on the donor-specific suppressive ability of Treg.
- Method 1 Spleen cells are collected from Foxp3/hCD2-KI C57BL6 mice (hCD2-KI mice) and adjusted to 4 x 10 6 cells/mL. 2.
- Spleen cells from Balb/c mice are collected and irradiated with 30 Gy of X-rays, then adjusted to a concentration of 4 x 10 6 cells/mL and mixed with hCD2-KI mouse spleen cells at a 1:1 ratio.
- Anti-IL-2 mAb final concentration: 10 ⁇ g/mL
- the cells are seeded onto a 10 cm dish, and cultured at 37° C.
- B6 mouse spleen cells, LPS-stimulated Balb/c mouse spleen cells or LPS-stimulated CBA/Nslc mouse spleen cells, and the cultured whole cells or isolated Tregs from 5. are prepared to the concentrations shown in Table 1 and seeded on a U-bottom 96-well plate. 7. Culture in a 37°C, 5% CO2 incubator for 3 days.
- IL-2 Measurement Two types of commercially available human peripheral blood mononuclear cells (LONZA CC-2702) were used as donor and recipient cells.
- the donor cells (Batch#22TL290862) were suspended in 15mL of RPMI medium (FUJIFILM Wako Pure Chemical Industries, Ltd.) containing 10% FCS (SELBORNE), and then irradiated with 25Gy of radiation.
- anti-CD80 antibody (manufactured by JUNTEN BIO Co., Ltd.) was added to the donor cells and recipient cells (Batch#23TL043676) at a final concentration of 5 mg/L, respectively, and then allowed to stand on ice for 30 minutes.
- donor cells and recipient cells were collected by centrifugation, and CD86 antibody (manufactured by JUNTEN BIO Co., Ltd.) was added to the donor cells at a final concentration of 5 mg/L.
- donor cells and recipient cells were prepared to a final concentration of 5.0x10 ⁇ 7 cells/20mL in two T flasks (Thermo scientific, 25 cm2 ) to which 20mL of 10% FCS-containing RPMI medium had been added.
- Anti-CD80 antibody (manufactured by JUNTEN BIO Co., Ltd.) and anti-CD86 antibody (manufactured by JUNTEN BIO Co., Ltd.) were added to one T flask to a final concentration of 5mg/L, and the remaining one was used as a negative control without adding any antibody.
- the two T flasks were placed in a 5% CO2 incubator set at 37°C.
- the culture supernatant was collected before the start of the culture and on days 1, 2, 3, 4, and 7 after the start of the culture, and the IL-2 concentration was measured by the Cytometric Bead Array (CBA) method.
- CBA Cytometric Bead Array
- the cells cultured for 7 days were added to a co-culture system of donor cells (Batch #22 TL290862: 2.0x10 ⁇ 5 cells/well) and recipient cells (Batch #23 TL043676: 2.0x10 ⁇ 5 cells/well) irradiated with 25 Gy, and the concentrations of IFN- ⁇ and granzyme B in the culture supernatant 5 days after culturing in a 96-well plate (CORNING) were measured by the CBA method. The results are shown in Table 4. By adding cells cultured in the presence of anti-CD80 and anti-CD86 antibodies, the concentrations of IFN- ⁇ and granzyme B were reduced in a cell concentration-dependent manner, confirming that MLR was suppressed.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Immunology (AREA)
- Chemical & Material Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Public Health (AREA)
- Organic Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Biomedical Technology (AREA)
- Cell Biology (AREA)
- Zoology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Biotechnology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Microbiology (AREA)
- Wood Science & Technology (AREA)
- Genetics & Genomics (AREA)
- Epidemiology (AREA)
- Pulmonology (AREA)
- General Engineering & Computer Science (AREA)
- Biochemistry (AREA)
- Transplantation (AREA)
- Mycology (AREA)
- Hematology (AREA)
- Developmental Biology & Embryology (AREA)
- Virology (AREA)
- Peptides Or Proteins (AREA)
Abstract
The present disclosure provides a method for selectively inducing antigen-specific inductive regulatory T cells from cells derived from a subject. Specifically, in one embodiment, the present disclosure provides a method for selectively inducing antigen-specific inductive regulatory T cells from cells derived from a subject. The method includes a step for mixing: a factor with respect to selectively inducing antigen-specific inductive regulatory T cells; cells derived from the subject; and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen not derived from the subject.
Description
肝臓移植は、末期の肝不全の患者に対する最終的な処置として広く使用されてきた。毎年、海外では20,000の症例があり、日本では500を超える症例がある。
Liver transplantation has been widely used as a definitive treatment for patients with end-stage liver failure. There are 20,000 cases overseas and more than 500 cases in Japan each year.
移植は、末期の腎臓、心臓、肝臓、及び膵臓の臓器不全に対する選択の主な処置の1つであり、近年における移植拒絶反応の処置の著しい進歩にもかかわらず、移植の大部分は最終的には拒絶される。連続的な薬物療法に依存する現在の免疫抑制レジメンは、薬物までもが移植に対して明確に向けられた反応を阻害することができないので、臓器移植患者に、感染症や癌に対する感受性の増加を生じやすくしてしまう。
Transplantation is one of the primary treatments of choice for end-stage renal, cardiac, hepatic, and pancreatic organ failure, and despite significant advances in the treatment of transplant rejection in recent years, the majority of transplants are ultimately rejected. Current immunosuppressive regimens, which rely on continuous drug therapy, predispose organ transplant patients to increased susceptibility to infections and cancer, as even the drugs fail to inhibit responses specifically directed against the transplant.
免疫寛容を誘導する技術として、T細胞の抗原特異的な免疫不応答(アナジー)の誘導がある。IL-2存在下で制御性T細胞(Treg)をドナー細胞と共培養し、分裂増殖させたドナー特異的Tregを治療に用いる方法や、低用量のIL-2を投与し生体内でTregを増殖させ、治療に用いる方法が存在する。しかし、これらの方法には、IL-2による他のT細胞活性化のリスク、ドナー特異的ではないポリクローナルなTregの増殖等の問題点がある。
One technique for inducing immune tolerance is the induction of antigen-specific immune unresponsiveness (anergy) in T cells. One method involves co-culturing regulatory T cells (Treg) with donor cells in the presence of IL-2, and using the donor-specific Tregs that have been divided and proliferated for treatment, while another method involves administering a low dose of IL-2 to proliferate Tregs in vivo and using them for treatment. However, these methods have problems, such as the risk of IL-2 activating other T cells and the proliferation of polyclonal Tregs that are not donor-specific.
本発明者らは、上記課題を解決するために、特定の因子を細胞と接触させることにより、抗原特異的誘導型抑制性T細胞を選択的に誘導させることを新たに見出した。さらに、本発明者らは、抗原特異的誘導型抑制性T細胞の誘導において、IL-2の産生または機能を調節することが重要であることを見出し、IL-2の産生または機能を調節することによる新たな抗原特異的誘導型抑制性T細胞の誘導方法も見出した。
In order to solve the above problems, the inventors have newly discovered that antigen-specific induced suppressor T cells can be selectively induced by contacting cells with a specific factor. Furthermore, the inventors have discovered that regulating the production or function of IL-2 is important in inducing antigen-specific induced suppressor T cells, and have also discovered a new method for inducing antigen-specific induced suppressor T cells by regulating the production or function of IL-2.
したがって、本願発明は、例えば、以下を提供する。
(項目1)被験体由来の細胞から抗原特異的誘導型抑制性T細胞を選択的に誘導させる方法であって、抗原特異的誘導型抑制性T細胞を選択的に誘導する因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該被験体に由来しない抗原の含有物とを混合する工程を含む、方法。
(項目2)前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子である、上記項目に記載の方法。
(項目3)前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、IL-2の産生を阻害することができる阻害因子または産生されたIL-2の機能を阻害することができる阻害因子である、上記項目のいずれか一項に記載の方法。
(項目4)前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、IL-2とIL-2受容体(IL-2R)との相互作用を阻害することができる阻害因子、ならびに/あるいは、CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子である、上記項目のいずれか一項に記載の方法。
(項目5)前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、抗IL-2抗体、抗IL-2R抗体、抗IL-2Rα(CD25)抗体、抗IL-2Rβ(CD122)抗体またはこれらの抗原結合断片から選択される前記IL-2とIL-2Rとの相互作用を阻害することができる阻害因子である、上記項目のいずれか一項に記載の方法。
(項目6)前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、抗CD80抗体、抗CD86抗体、CD80およびCD86に対する二重特異性抗体、抗CD28抗体もしくはこれらの抗原結合断片、CTLA4-Ig融合タンパク質、およびCD28-Ig融合タンパク質からなる群より選択される前記CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子である、上記項目のいずれか一項に記載の方法。
(項目7)被験体由来の細胞から誘導型抑制性T細胞を作製する方法であって、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合する工程を含む、方法。
(項目8)前記IL-2の産生を調節することができる調節因子が、CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子、ならびにIL-2産生を阻害する小分子化合物から選択される、上記項目のいずれか一項に記載の方法。
(項目9)前記産生されたIL-2の機能を調節することができる調節因子が、IL-2とIL-2受容体(IL-2R)との相互作用を阻害することができる阻害因子、およびIL-2の作用を阻害する小分子化合物から選択される、上記項目のいずれか一項に記載の方法。
(項目10)前記CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子は、抗CD80抗体、抗CD86抗体、CD80およびCD86に対する二重特異性抗体、抗CD28抗体もしくはこれらの抗原結合断片、CTLA4-Ig融合タンパク質、およびCD28-Ig融合タンパク質からなる群より選択される、上記項目のいずれか一項に記載の方法。
(項目11)前記IL-2とIL-2受容体(IL-2R)との相互作用を阻害することができる阻害因子が、抗IL-2抗体、抗IL-2R抗体、抗IL-2Rα(CD25)抗体、抗IL-2Rβ(CD122)抗体またはこれらの抗原結合断片から選択される、上記項目のいずれか一項に記載の方法。
(項目12)被験体由来の細胞から抗原特異的誘導型抑制性T細胞を選択的に誘導させるための組成物であって、該組成物は、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子を含み、該組成物が、該被験体由来の細胞、および該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物と接触させられることを特徴とする、組成物。
(項目12A)被験体由来の細胞から抗原特異的誘導型抑制性T細胞を選択的に誘導させるための、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子であって、該調節因子が、該被験体由来の細胞、および該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物と接触させられることを特徴とする、調節因子。
(項目13)免疫応答により媒介される被験体の疾患を処置するための組成物であって、該組成物が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合することによって選択的に誘導された抗原特異的誘導型抑制性T細胞を含む、組成物。
(項目13A)免疫応答により媒介される被験体の疾患を処置する方法であって、該方法が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合する工程と、該混合によって選択的に誘導された抗原特異的誘導型抑制性T細胞を該被験体に投与する工程と、を含む、方法。
(項目13B)免疫応答により媒介される被験体の疾患を処置するための、抗原特異的誘導型抑制性T細胞であって、該抗原特異的誘導型抑制性T細胞が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合することによって選択的に誘導される、抗原特異的誘導型抑制性T細胞。
(項目13C)免疫応答により媒介される被験体の疾患を処置するための医薬の製造における、抗原特異的誘導型抑制性T細胞の使用であって、該抗原特異的誘導型抑制性T細胞が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合することによって選択的に誘導される、使用。
(項目14)前記疾患が、移植免疫拒絶反応、アレルギー、自己免疫疾患、移植片対宿主病、およびiPS細胞もしくはES細胞またはそれらから分化した細胞、組織もしくは臓器の移植によって引き起こされる免疫拒絶反応からなる群より選択される、上記項目のいずれか一項に記載の組成物、上記項目のいずれか一項に記載の方法、上記項目のいずれか一項に記載の抗原特異的誘導型抑制性T細胞、または上記項目のいずれか一項に記載の使用。
(項目15)前記被験体はヒトである、上記項目のいずれか一項に記載の組成物、方法、抗原特異的誘導型抑制性T細胞、または使用。 Thus, the present invention provides, for example:
(Item 1) A method for selectively inducing antigen-specific induced inhibitory T cells from cells derived from a subject, the method comprising a step of mixing a factor that selectively induces antigen-specific induced inhibitory T cells, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a material containing an antigen not derived from the subject.
(Item 2) The method according to the above item, wherein the factor that selectively induces antigen-specific induced suppressor T cells is a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2.
(Item 3) The method according to any one of the above items, wherein the factor that selectively induces antigen-specific induced suppressor T cells is an inhibitory factor capable of inhibiting the production of IL-2 or an inhibitory factor capable of inhibiting the function of the produced IL-2.
(Item 4) The method according to any one of the above items, wherein the factor that selectively induces antigen-specific induced suppressor T cells is an inhibitory factor capable of inhibiting the interaction between IL-2 and IL-2 receptor (IL-2R) and/or an inhibitory factor capable of inhibiting the interaction between CD80 and/or CD86 and CD28.
(Item 5) The method according to any one of the above items, wherein the factor that selectively induces antigen-specific induced inhibitory T cells is an inhibitor capable of inhibiting the interaction between IL-2 and IL-2R, selected from an anti-IL-2 antibody, an anti-IL-2R antibody, an anti-IL-2Rα (CD25) antibody, an anti-IL-2Rβ (CD122) antibody, or an antigen-binding fragment thereof.
(Item 6) The method according to any one of the above items, wherein the factor that selectively induces antigen-specific induced inhibitory T cells is an inhibitor capable of inhibiting the interaction between CD80 and/or CD86 and CD28, selected from the group consisting of an anti-CD80 antibody, an anti-CD86 antibody, a bispecific antibody against CD80 and CD86, an anti-CD28 antibody or an antigen-binding fragment thereof, a CTLA4-Ig fusion protein, and a CD28-Ig fusion protein.
(Item 7) A method for producing induced suppressor T cells from cells derived from a subject, the method comprising the step of mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
(Item 8) The method according to any one of the above items, wherein the regulatory factor capable of regulating the production of IL-2 is selected from an inhibitor capable of inhibiting the interaction of CD80 and/or CD86 with CD28, and a small molecule compound that inhibits IL-2 production.
(Item 9) The method according to any one of the above items, wherein the regulator capable of regulating the function of the produced IL-2 is selected from an inhibitor capable of inhibiting the interaction of IL-2 with IL-2 receptor (IL-2R) and a small molecule compound that inhibits the action of IL-2.
(Item 10) The method of any one of the above items, wherein the inhibitor capable of inhibiting the interaction of CD80 and/or CD86 with CD28 is selected from the group consisting of an anti-CD80 antibody, an anti-CD86 antibody, a bispecific antibody against CD80 and CD86, an anti-CD28 antibody or an antigen-binding fragment thereof, a CTLA4-Ig fusion protein, and a CD28-Ig fusion protein.
(Item 11) The method according to any one of the above items, wherein the inhibitor capable of inhibiting the interaction between IL-2 and IL-2 receptor (IL-2R) is selected from an anti-IL-2 antibody, an anti-IL-2R antibody, an anti-IL-2Rα (CD25) antibody, an anti-IL-2Rβ (CD122) antibody, or an antigen-binding fragment thereof.
(Item 12) A composition for selectively inducing antigen-specific induced suppressor T cells from cells derived from a subject, the composition comprising a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, characterized in that the composition is contacted with cells derived from the subject and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
(Item 12A) A regulatory agent capable of regulating the production of IL-2 or the function of produced IL-2 in order to selectively induce antigen-specific induced suppressor T cells from cells derived from a subject, characterized in that the regulatory agent is contacted with cells derived from the subject and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
(Item 13) A composition for treating a disease in a subject mediated by an immune response, the composition comprising a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, and antigen-specific induced suppressor T cells selectively induced by mixing cells derived from the subject with an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
(Item 13A) A method for treating a disease in a subject mediated by an immune response, the method comprising the steps of mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen, and administering to the subject antigen-specific induced suppressor T cells selectively induced by the mixing.
(Item 13B) An antigen-specific induced suppressor T cell for treating a disease in a subject mediated by an immune response, the antigen-specific induced suppressor T cell being selectively induced by mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
(Item 13C) Use of antigen-specific induced inhibitory T cells in the manufacture of a medicine for treating a disease in a subject mediated by an immune response, wherein the antigen-specific induced inhibitory T cells are selectively induced by mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
(Item 14) The composition described in any one of the above items, the method described in any one of the above items, the antigen-specific induced suppressor T cell described in any one of the above items, or the use described in any one of the above items, wherein the disease is selected from the group consisting of transplant immune rejection, allergy, autoimmune disease, graft-versus-host disease, and immune rejection caused by transplantation of iPS cells or ES cells, or cells, tissues, or organs differentiated therefrom.
(Item 15) The composition, method, antigen-specific induced suppressor T cell, or use described in any one of the above items, wherein the subject is a human.
(項目1)被験体由来の細胞から抗原特異的誘導型抑制性T細胞を選択的に誘導させる方法であって、抗原特異的誘導型抑制性T細胞を選択的に誘導する因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該被験体に由来しない抗原の含有物とを混合する工程を含む、方法。
(項目2)前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子である、上記項目に記載の方法。
(項目3)前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、IL-2の産生を阻害することができる阻害因子または産生されたIL-2の機能を阻害することができる阻害因子である、上記項目のいずれか一項に記載の方法。
(項目4)前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、IL-2とIL-2受容体(IL-2R)との相互作用を阻害することができる阻害因子、ならびに/あるいは、CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子である、上記項目のいずれか一項に記載の方法。
(項目5)前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、抗IL-2抗体、抗IL-2R抗体、抗IL-2Rα(CD25)抗体、抗IL-2Rβ(CD122)抗体またはこれらの抗原結合断片から選択される前記IL-2とIL-2Rとの相互作用を阻害することができる阻害因子である、上記項目のいずれか一項に記載の方法。
(項目6)前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、抗CD80抗体、抗CD86抗体、CD80およびCD86に対する二重特異性抗体、抗CD28抗体もしくはこれらの抗原結合断片、CTLA4-Ig融合タンパク質、およびCD28-Ig融合タンパク質からなる群より選択される前記CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子である、上記項目のいずれか一項に記載の方法。
(項目7)被験体由来の細胞から誘導型抑制性T細胞を作製する方法であって、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合する工程を含む、方法。
(項目8)前記IL-2の産生を調節することができる調節因子が、CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子、ならびにIL-2産生を阻害する小分子化合物から選択される、上記項目のいずれか一項に記載の方法。
(項目9)前記産生されたIL-2の機能を調節することができる調節因子が、IL-2とIL-2受容体(IL-2R)との相互作用を阻害することができる阻害因子、およびIL-2の作用を阻害する小分子化合物から選択される、上記項目のいずれか一項に記載の方法。
(項目10)前記CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子は、抗CD80抗体、抗CD86抗体、CD80およびCD86に対する二重特異性抗体、抗CD28抗体もしくはこれらの抗原結合断片、CTLA4-Ig融合タンパク質、およびCD28-Ig融合タンパク質からなる群より選択される、上記項目のいずれか一項に記載の方法。
(項目11)前記IL-2とIL-2受容体(IL-2R)との相互作用を阻害することができる阻害因子が、抗IL-2抗体、抗IL-2R抗体、抗IL-2Rα(CD25)抗体、抗IL-2Rβ(CD122)抗体またはこれらの抗原結合断片から選択される、上記項目のいずれか一項に記載の方法。
(項目12)被験体由来の細胞から抗原特異的誘導型抑制性T細胞を選択的に誘導させるための組成物であって、該組成物は、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子を含み、該組成物が、該被験体由来の細胞、および該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物と接触させられることを特徴とする、組成物。
(項目12A)被験体由来の細胞から抗原特異的誘導型抑制性T細胞を選択的に誘導させるための、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子であって、該調節因子が、該被験体由来の細胞、および該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物と接触させられることを特徴とする、調節因子。
(項目13)免疫応答により媒介される被験体の疾患を処置するための組成物であって、該組成物が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合することによって選択的に誘導された抗原特異的誘導型抑制性T細胞を含む、組成物。
(項目13A)免疫応答により媒介される被験体の疾患を処置する方法であって、該方法が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合する工程と、該混合によって選択的に誘導された抗原特異的誘導型抑制性T細胞を該被験体に投与する工程と、を含む、方法。
(項目13B)免疫応答により媒介される被験体の疾患を処置するための、抗原特異的誘導型抑制性T細胞であって、該抗原特異的誘導型抑制性T細胞が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合することによって選択的に誘導される、抗原特異的誘導型抑制性T細胞。
(項目13C)免疫応答により媒介される被験体の疾患を処置するための医薬の製造における、抗原特異的誘導型抑制性T細胞の使用であって、該抗原特異的誘導型抑制性T細胞が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合することによって選択的に誘導される、使用。
(項目14)前記疾患が、移植免疫拒絶反応、アレルギー、自己免疫疾患、移植片対宿主病、およびiPS細胞もしくはES細胞またはそれらから分化した細胞、組織もしくは臓器の移植によって引き起こされる免疫拒絶反応からなる群より選択される、上記項目のいずれか一項に記載の組成物、上記項目のいずれか一項に記載の方法、上記項目のいずれか一項に記載の抗原特異的誘導型抑制性T細胞、または上記項目のいずれか一項に記載の使用。
(項目15)前記被験体はヒトである、上記項目のいずれか一項に記載の組成物、方法、抗原特異的誘導型抑制性T細胞、または使用。 Thus, the present invention provides, for example:
(Item 1) A method for selectively inducing antigen-specific induced inhibitory T cells from cells derived from a subject, the method comprising a step of mixing a factor that selectively induces antigen-specific induced inhibitory T cells, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a material containing an antigen not derived from the subject.
(Item 2) The method according to the above item, wherein the factor that selectively induces antigen-specific induced suppressor T cells is a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2.
(Item 3) The method according to any one of the above items, wherein the factor that selectively induces antigen-specific induced suppressor T cells is an inhibitory factor capable of inhibiting the production of IL-2 or an inhibitory factor capable of inhibiting the function of the produced IL-2.
(Item 4) The method according to any one of the above items, wherein the factor that selectively induces antigen-specific induced suppressor T cells is an inhibitory factor capable of inhibiting the interaction between IL-2 and IL-2 receptor (IL-2R) and/or an inhibitory factor capable of inhibiting the interaction between CD80 and/or CD86 and CD28.
(Item 5) The method according to any one of the above items, wherein the factor that selectively induces antigen-specific induced inhibitory T cells is an inhibitor capable of inhibiting the interaction between IL-2 and IL-2R, selected from an anti-IL-2 antibody, an anti-IL-2R antibody, an anti-IL-2Rα (CD25) antibody, an anti-IL-2Rβ (CD122) antibody, or an antigen-binding fragment thereof.
(Item 6) The method according to any one of the above items, wherein the factor that selectively induces antigen-specific induced inhibitory T cells is an inhibitor capable of inhibiting the interaction between CD80 and/or CD86 and CD28, selected from the group consisting of an anti-CD80 antibody, an anti-CD86 antibody, a bispecific antibody against CD80 and CD86, an anti-CD28 antibody or an antigen-binding fragment thereof, a CTLA4-Ig fusion protein, and a CD28-Ig fusion protein.
(Item 7) A method for producing induced suppressor T cells from cells derived from a subject, the method comprising the step of mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
(Item 8) The method according to any one of the above items, wherein the regulatory factor capable of regulating the production of IL-2 is selected from an inhibitor capable of inhibiting the interaction of CD80 and/or CD86 with CD28, and a small molecule compound that inhibits IL-2 production.
(Item 9) The method according to any one of the above items, wherein the regulator capable of regulating the function of the produced IL-2 is selected from an inhibitor capable of inhibiting the interaction of IL-2 with IL-2 receptor (IL-2R) and a small molecule compound that inhibits the action of IL-2.
(Item 10) The method of any one of the above items, wherein the inhibitor capable of inhibiting the interaction of CD80 and/or CD86 with CD28 is selected from the group consisting of an anti-CD80 antibody, an anti-CD86 antibody, a bispecific antibody against CD80 and CD86, an anti-CD28 antibody or an antigen-binding fragment thereof, a CTLA4-Ig fusion protein, and a CD28-Ig fusion protein.
(Item 11) The method according to any one of the above items, wherein the inhibitor capable of inhibiting the interaction between IL-2 and IL-2 receptor (IL-2R) is selected from an anti-IL-2 antibody, an anti-IL-2R antibody, an anti-IL-2Rα (CD25) antibody, an anti-IL-2Rβ (CD122) antibody, or an antigen-binding fragment thereof.
(Item 12) A composition for selectively inducing antigen-specific induced suppressor T cells from cells derived from a subject, the composition comprising a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, characterized in that the composition is contacted with cells derived from the subject and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
(Item 12A) A regulatory agent capable of regulating the production of IL-2 or the function of produced IL-2 in order to selectively induce antigen-specific induced suppressor T cells from cells derived from a subject, characterized in that the regulatory agent is contacted with cells derived from the subject and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
(Item 13) A composition for treating a disease in a subject mediated by an immune response, the composition comprising a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, and antigen-specific induced suppressor T cells selectively induced by mixing cells derived from the subject with an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
(Item 13A) A method for treating a disease in a subject mediated by an immune response, the method comprising the steps of mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen, and administering to the subject antigen-specific induced suppressor T cells selectively induced by the mixing.
(Item 13B) An antigen-specific induced suppressor T cell for treating a disease in a subject mediated by an immune response, the antigen-specific induced suppressor T cell being selectively induced by mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
(Item 13C) Use of antigen-specific induced inhibitory T cells in the manufacture of a medicine for treating a disease in a subject mediated by an immune response, wherein the antigen-specific induced inhibitory T cells are selectively induced by mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
(Item 14) The composition described in any one of the above items, the method described in any one of the above items, the antigen-specific induced suppressor T cell described in any one of the above items, or the use described in any one of the above items, wherein the disease is selected from the group consisting of transplant immune rejection, allergy, autoimmune disease, graft-versus-host disease, and immune rejection caused by transplantation of iPS cells or ES cells, or cells, tissues, or organs differentiated therefrom.
(Item 15) The composition, method, antigen-specific induced suppressor T cell, or use described in any one of the above items, wherein the subject is a human.
本開示において、上記1または複数の特徴は、明示された組み合わせに加え、さらに組み合わせて提供されうることが意図される。本開示のなおさらなる実施形態および利点は、必要に応じて以下の詳細な説明を読んで理解すれば、当業者に認識される。
It is contemplated that in the present disclosure, one or more of the above features may be provided in combinations in addition to those combinations explicitly stated. Still further embodiments and advantages of the present disclosure will be recognized by those skilled in the art upon reading and understanding the following detailed description, if necessary.
本開示の方法は、特定の因子を細胞と接触させることにより、抗原特異的誘導型抑制性T細胞を選択的に誘導させることが可能である。
The method disclosed herein makes it possible to selectively induce antigen-specific induced suppressor T cells by contacting cells with a specific factor.
以下、本開示を最良の形態を示しながら説明する。本明細書の全体にわたり、単数形の表現は、特に言及しない限り、その複数形の概念をも含むことが理解されるべきである。従って、単数形の冠詞(例えば、英語の場合は「a」、「an」、「the」など)は、特に言及しない限り、その複数形の概念をも含むことが理解されるべきである。また、本明細書において使用される用語は、特に言及しない限り、当該分野で通常用いられる意味で用いられることが理解されるべきである。したがって、他に定義されない限り、本明細書中で使用されるすべての専門用語および科学技術用語は、本開示の属する分野の当業者によって一般的に理解されるのと同じ意味を有する。矛盾する場合、本明細書(定義を含めて)が優先する。
The present disclosure will be described below while showing the best mode. Throughout this specification, singular expressions should be understood to include the concept of the plural, unless otherwise specified. Thus, singular articles (e.g., in the case of English, "a," "an," "the," etc.) should be understood to include the concept of the plural, unless otherwise specified. In addition, terms used in this specification should be understood to be used in the sense commonly used in the field, unless otherwise specified. Thus, unless otherwise defined, all technical and scientific terms used in this specification have the same meaning as commonly understood by those skilled in the art to which this disclosure belongs. In case of conflict, the present specification (including definitions) will take precedence.
(用語の定義)
本明細書において「約」とは、本明細書で使用される場合、後に続く数値の±10%を意味する。 (Definition of terms)
As used herein, "about" means ±10% of the numerical value that follows.
本明細書において「約」とは、本明細書で使用される場合、後に続く数値の±10%を意味する。 (Definition of terms)
As used herein, "about" means ±10% of the numerical value that follows.
本明細書において、「免疫寛容」とは、特定の抗原に対する特異的免疫反応を示さないか、特異的免疫反応が抑制された状態のことである。免疫寛容は、免疫細胞(特にT細胞)が特定の抗原に対する特異的免疫反応を示さないか、特異的免疫反応が抑制された状態と、ヒトが特定の抗原に対する特異的免疫反応を示さないか、特異的免疫反応が抑制された状態の両方またはいずれか一方を意味していてもよい。免疫寛容が惹起されることにより、免疫拒絶反応に対する処置が可能になったり、アレルギーに対する治療が可能なったりすることから注目されている。本明細書において、「アナジー」とは、抗原提示細胞から抗原を提示される際に共刺激が入力されないことにより、当該次回に共刺激のある条件で刺激されても反応できなくなった状態を意味する。したがって、本明細書では「アナジー細胞」とは、免疫寛容が生じた(免疫不応答の)細胞をいい、「アナジーT細胞」は、免疫寛容が生じた(免疫不応答の)T細胞であり、活性化されないことのほか、再び同じ抗原にであったときに無反応であるT細胞も包含される。なお、本明細書において、「免疫寛容を誘導されたPBMC(またはT細胞)」と「アナジーなPBMC(またはT細胞)」とは同意義である。CD44陽性を確認することによって確認することができるが、これに限定されない。
In this specification, "immune tolerance" refers to a state in which a specific immune response to a specific antigen is not shown or the specific immune response is suppressed. Immune tolerance may mean both or either of a state in which immune cells (especially T cells) do not show a specific immune response to a specific antigen or the specific immune response is suppressed, and a state in which a human does not show a specific immune response to a specific antigen or the specific immune response is suppressed. Immune tolerance has attracted attention because it makes it possible to treat immune rejection reactions and allergies by inducing immune tolerance. In this specification, "anergy" refers to a state in which no costimulation is input when an antigen is presented from an antigen-presenting cell, and therefore the cell is unable to respond even if stimulated under costimulatory conditions the next time. Therefore, in this specification, "anergy cells" refer to cells in which immune tolerance has occurred (immune unresponsive), and "anergy T cells" refer to T cells in which immune tolerance has occurred (immune unresponsive), and include T cells that are not activated and are unresponsive when they encounter the same antigen again. In this specification, "PBMCs (or T cells) in which immune tolerance has been induced" and "anergy PBMCs (or T cells)" have the same meaning. They can be confirmed by, but are not limited to, confirming that they are CD44 positive.
本明細書において、「被験体」とは、飼育動物(例えば、ウシ、ヒツジ、ネコ、イヌ、ウマ)、霊長類(例えば、ヒト、およびサルなどの非ヒト霊長類)、ウサギ、ならびに、げっ歯類(例えば、マウスおよびラット)を含む。特定の実施形態では、被験体は、ヒトである。
As used herein, "subject" includes domestic animals (e.g., cows, sheep, cats, dogs, horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats). In certain embodiments, the subject is a human.
本明細書において「因子」とは、意図する目的を達成することができる限りどのような物質または他の要素(例えば、光、放射能、熱、電気などのエネルギー)でもあってもよい。そのような物質としては、例えば、タンパク質、ポリペプチド、オリゴペプチド、ペプチド、ポリヌクレオチド、オリゴヌクレオチド、ヌクレオチド、核酸(例えば、cDNA、ゲノムDNAのようなDNA、mRNAのようなRNAを含む)、ポリサッカリド、オリゴサッカリド、脂質、有機低分子(例えば、ホルモン、リガンド、情報伝達物質、その他の有機低分子化合物、コンビナトリアルケミストリで合成された分子、医薬品として利用され得る低分子(例えば、低分子リガンドなど)など)、これらの複合分子が挙げられるがそれらに限定されない。
In this specification, the term "factor" may refer to any substance or other element (e.g., energy such as light, radioactivity, heat, electricity, etc.) as long as it can achieve the intended purpose. Examples of such substances include, but are not limited to, proteins, polypeptides, oligopeptides, peptides, polynucleotides, oligonucleotides, nucleotides, nucleic acids (e.g., DNA such as cDNA and genomic DNA, and RNA such as mRNA), polysaccharides, oligosaccharides, lipids, small organic molecules (e.g., hormones, ligands, signaling substances, other small organic compounds, molecules synthesized by combinatorial chemistry, small molecules that can be used as pharmaceuticals (e.g., small molecule ligands, etc.)), and composite molecules thereof.
本明細書において「調節因子」は、所定の作用(例えば、相互作用、シグナル伝達、タンパク質産生、タンパク質機能など)を正または負の調節をするあらゆる種類の低分子、タンパク質、核酸、脂質、糖などの因子をいう。したがって、調節は亢進(増加)ならびに低下(阻害、抑制)および消失等の任意のレベルへの変更または変更させないことを含み、本明細書では「調節因子」は「阻害因子」および「亢進因子」等を含む。特定の理論に束縛されることは望まないが、IL-2の産生を調節するか、産生されたIL-2の機能を調節することによって、特定の抗原に対して特異的な誘導型抑制性T細胞の増殖を促進する。IL-2は、特定の抗原に対して非特異的な誘導型抑制性T細胞の増殖も促進するため、IL-2の産生を調節するか、産生されたIL-2の機能を調節することによって、非特異的な誘導型抑制性T細胞の増殖を抑制することが可能である。IL-2の産生および機能を完全に阻害すると抗原特異的誘導型抑制性T細胞の増殖も抑制してしまうため、所定量のIL-2を存在することが好ましい。本開示の方法、因子、および組成物は、所定量のIL-2の存在を達成するように調節することが可能である。
As used herein, the term "regulatory factor" refers to any type of factor, such as a small molecule, protein, nucleic acid, lipid, or sugar, that positively or negatively regulates a given action (e.g., interaction, signal transduction, protein production, protein function, etc.). Thus, regulation includes modification or non-modification to any level, such as enhancement (increase), as well as reduction (inhibition, suppression), and disappearance, and as used herein, "regulatory factor" includes "inhibitory factor" and "enhancing factor", etc. Although not wishing to be bound by a particular theory, the proliferation of induced suppressive T cells specific to a particular antigen is promoted by regulating the production of IL-2 or the function of the produced IL-2. Since IL-2 also promotes the proliferation of induced suppressive T cells nonspecific to a particular antigen, it is possible to suppress the proliferation of nonspecific induced suppressive T cells by regulating the production of IL-2 or the function of the produced IL-2. Since complete inhibition of the production and function of IL-2 also suppresses the proliferation of antigen-specific induced suppressive T cells, it is preferable to have a certain amount of IL-2. The methods, factors, and compositions disclosed herein can be adjusted to achieve the presence of a predetermined amount of IL-2.
したがって、本明細書において「阻害因子」は、所定の作用(例えば、相互作用、シグナル伝達、タンパク質産生、タンパク質機能など)を阻害することができる、あらゆる種類の低分子、タンパク質、核酸、脂質、糖などの因子をいう。特定の理論に束縛されることは望まないが、本開示において、細胞表面のCD80および/またはCD86と、CD28との相互作用をブロックし、CD28共刺激シグナルを阻害することにより、T細胞にアナジーを誘導する。本開示では、CD80および/またはCD86と、CD28との相互作用をブロックするのに使用される阻害因子は、低分子、タンパク質、核酸、脂質、糖、およびこれらの組み合わせからなる群から選択される。一局面では、上記タンパク質は、抗体もしくはその改変体、または細胞表面分子もしくはその改変体である。別の局面では、上記抗体の改変体は、抗原結合断片である。別の局面では、上記細胞表面分子の改変体は、融合タンパク質である。別の局面では、上記阻害因子は、抗CD80抗体、抗CD86抗体、CD80およびCD86に対する二重特異性抗体、抗CD28抗体もしくはこれらの抗原結合断片、CTLA4-Ig融合タンパク質、CD28-Ig融合タンパク質からなる群より選択される。別の局面では、上記CTLA4-Ig融合タンパク質は、アバタセプトまたはベラタセプトである。また、上記相互作用を間接的に阻害する因子(例えば、シグナル伝達の上流または下流のシグナルの阻害因子)も組み合わせて使用することも想定される。
Therefore, as used herein, the term "inhibitory factor" refers to any type of factor, such as a small molecule, protein, nucleic acid, lipid, or sugar, that can inhibit a certain action (e.g., interaction, signal transduction, protein production, protein function, etc.). Without wishing to be bound by a particular theory, in the present disclosure, anergy is induced in T cells by blocking the interaction between CD80 and/or CD86 on the cell surface and CD28 and inhibiting the CD28 costimulatory signal. In the present disclosure, the inhibitor used to block the interaction between CD80 and/or CD86 and CD28 is selected from the group consisting of a small molecule, a protein, a nucleic acid, a lipid, a sugar, and a combination thereof. In one aspect, the protein is an antibody or a variant thereof, or a cell surface molecule or a variant thereof. In another aspect, the variant of the antibody is an antigen-binding fragment. In another aspect, the variant of the cell surface molecule is a fusion protein. In another aspect, the inhibitor is selected from the group consisting of an anti-CD80 antibody, an anti-CD86 antibody, a bispecific antibody against CD80 and CD86, an anti-CD28 antibody or an antigen-binding fragment thereof, a CTLA4-Ig fusion protein, and a CD28-Ig fusion protein. In another aspect, the CTLA4-Ig fusion protein is abatacept or belatacept. It is also envisioned that an inhibitor that indirectly inhibits the above interaction (e.g., an inhibitor of an upstream or downstream signal transduction) may also be used in combination.
本明細書において広義の「抗体」は、抗原上の特定のエピトープに特異的に結合することができる分子またはその集団をいう。本明細書における広義の「抗体」は、全長抗体(すなわち、Fc部分を有する抗体)であっても、Fc部分を欠いている抗体であってもよい。Fc部分を欠いている抗体は、目的の抗原に結合することができればよく、そのような抗体としては、例えば、Fab抗体、F(ab’)2抗体、Fab’抗体、Fv抗体、scFv抗体などが挙げられるが、これらに限定されない。抗体は、どのようなタイプの抗体であってもよく、すなわち、当該分野において公知の免疫グロブリンであってもよい。例示的な実施形態において、抗体は、アイソタイプIgA、IgD、IgE、IgG、またはIgMのクラスの抗体である。例示的な実施形態において、本明細書に記載される抗体は、1つまたは複数のアルファ、デルタ、イプシロン、ガンマ、および/またはミュー重鎖を含む。例示的な実施形態において、本明細書に記載される抗体は、1つまたは複数のカッパまたは軽鎖を含む。例示的な態様において、抗体は、IgG抗体であり、4つのヒトサブクラス:IgG1、IgG2、IgG3およびIgG4のうちの1つである。また、本開示における使用が想定される抗体としては、ラクダ科動物由来の抗体(例えば、VHH抗体)、サメ由来の抗体(例えば、一本鎖抗体)、ペプチボディ(peptibody)、ナノボディ(nanobody、単一ドメイン抗体)、ミニボディ(minibody)、多重特異性抗体(例えば、二重特異性抗体、ダイアボディ(diabody)、トリアボディ(triabody)、テトラボディ(tetrabody)、タンデムジ-scFV、タンデムトリ-scFv)などが挙げられ、これらは、当該分野において公知である。例えば、Korttら、Biomol Eng. 2001 18巻:95~108頁、(2001年)およびTodorovskaら、J Immunol Methods. 248巻:47~66頁、(2001年)などを参照されたい。また抗体は、抗体修飾物または抗体非修飾物を含む。抗体修飾物は、抗体と、例えばポリエチレングリコール等の各種分子が結合していてもよい。抗体修飾物は、抗体に公知の手法を用いて化学的な修飾を施すことによって得ることができる。人工的に作出された抗体および抗体の種々の修飾/改変方法については、生化学(2016)第88巻第3号380~385頁も参照のこと。
In the broad sense of the term "antibody" herein, a molecule or a group thereof that can specifically bind to a particular epitope on an antigen. In the broad sense of the term "antibody" herein, a full-length antibody (i.e., an antibody having an Fc portion) or an antibody lacking an Fc portion may be used. An antibody lacking an Fc portion may be capable of binding to an antigen of interest, and examples of such an antibody include, but are not limited to, a Fab antibody, an F(ab') 2 antibody, an Fab' antibody, an Fv antibody, and an scFv antibody. The antibody may be any type of antibody, i.e., an immunoglobulin known in the art. In an exemplary embodiment, the antibody is an antibody of the isotype IgA, IgD, IgE, IgG, or IgM class. In an exemplary embodiment, the antibody described herein comprises one or more alpha, delta, epsilon, gamma, and/or mu heavy chains. In an exemplary embodiment, the antibody described herein comprises one or more kappa or light chains. In exemplary aspects, the antibody is an IgG antibody and is one of the four human subclasses: IgG1, IgG2, IgG3, and IgG4. Antibodies contemplated for use in the present disclosure also include camelid-derived antibodies (e.g., VHH antibodies), shark-derived antibodies (e.g., single-chain antibodies), peptibodies, nanobodies (single domain antibodies), minibodies, multispecific antibodies (e.g., bispecific antibodies, diabodies, triabodies, tetrabodies, tandem di-scFv, tandem tri-scFv), and the like, which are known in the art. See, e.g., Kortt et al., Biomol Eng. 2001 18:95-108, (2001) and Todorovska et al., J Immunol Methods. 248:47-66, (2001). The antibody also includes modified or unmodified antibodies. The modified antibody may be bound to an antibody with various molecules such as polyethylene glycol. The modified antibody can be obtained by chemically modifying the antibody using a known method. For artificially produced antibodies and various methods of modifying/altering antibodies, see also Biochemistry (2016) 88:380-385.
本明細書において狭義の「抗体」は、抗原上の特定のエピトープに特異的に結合することができるイムノグロブリンまたはその集団をいい、その改変体を「抗体の改変体」という。本明細書における狭義の「抗体」は、全長抗体(すなわち、Fc部分を有する抗体)であり得、本明細書における「抗体の改変体」は、上記抗体のFc部分を欠いている改変体であり得る。したがって、本明細書において狭義の抗体は全長抗体とも称され得、抗体の改変体は全長抗体の改変体とも称され得る。Fc部分を欠いている改変体は、目的の抗原に結合することができればよく、そのような改変体としては、例えば、Fab抗体、F(ab’)2抗体、Fab’抗体、Fv抗体、scFv抗体などが挙げられるが、これらに限定されない。また抗体の改変体は、抗体修飾物または抗体非修飾物を含む。抗体修飾物は、抗体と、例えばポリエチレングリコール等の各種分子が結合していてもよい。抗体修飾物は、抗体に公知の手法を用いて化学的な修飾を施すことによって得ることができる。
In the present specification, the term "antibody" in the narrow sense refers to an immunoglobulin or a group thereof that can specifically bind to a specific epitope on an antigen, and a variant thereof is referred to as a "variant of an antibody". In the present specification, the term "antibody" in the narrow sense may be a full-length antibody (i.e., an antibody having an Fc portion), and in the present specification, the term "variant of an antibody" may be a variant lacking the Fc portion of the antibody. Thus, in the present specification, an antibody in the narrow sense may also be referred to as a full-length antibody, and a variant of an antibody may also be referred to as a variant of a full-length antibody. A variant lacking an Fc portion may be capable of binding to an antigen of interest, and examples of such variants include, but are not limited to, Fab antibodies, F(ab') 2 antibodies, Fab' antibodies, Fv antibodies, and scFv antibodies. In addition, variants of an antibody include modified or unmodified antibodies. The modified antibody may be bound to an antibody with various molecules such as polyethylene glycol. The modified antibody can be obtained by chemically modifying an antibody using a known method.
本開示の一実施形態において「ポリクローナル抗体」は、例えば、抗原に特異的なポリクローナル抗体の産生を誘導するために、哺乳類(例えば、ラット、マウス、ウサギ、ウシ、サル等)、鳥類等に、目的の抗原を含む免疫原を投与することによって生成することが可能である。免疫原の投与は、1つ以上の免疫剤、および所望の場合にはアジュバントの注入をしてもよい。アジュバントは、免疫応答を増加させるために使用されることもあり、フロイントアジュバント(完全または不完全)、ミネラルゲル(水酸化アルミニウム等)、または界面活性物質(リゾレシチン等)等を含んでいてもよい。免疫プロトコールは、当該技術分野で公知であり、選択する宿主生物に合わせて、免疫応答を誘発する任意の方法によって実施される場合がある(タンパク質実験ハンドブック,羊土社(2003):86-91.)。
In one embodiment of the present disclosure, a "polyclonal antibody" can be produced, for example, by administering an immunogen containing an antigen of interest to a mammal (e.g., rat, mouse, rabbit, cow, monkey, etc.), bird, etc., to induce production of polyclonal antibodies specific to the antigen. The administration of the immunogen may be by injection of one or more immunizing agents and, if desired, an adjuvant. Adjuvants may be used to increase the immune response and may include Freund's adjuvant (complete or incomplete), mineral gels (e.g., aluminum hydroxide), or surfactants (e.g., lysolecithin), etc. Immunization protocols are known in the art and may be performed by any method that induces an immune response, tailored to the host organism of choice (Protein Experiment Handbook, Yodosha (2003): 86-91.).
本開示の一実施形態において「モノクローナル抗体」は、集団を構成する個々の抗体が、少量自然に生じることが可能な突然変異を有する抗体を除いて、実質的に単一のエピトープに対応する同一な抗体である場合を含む。または、集団を構成する個々の抗体が、少量自然に生じることが可能な突然変異を有する抗体を除いて、実質的に同一である抗体であってもよい。モノクローナル抗体は高度に特異的であり、異なるエピトープに対応する異なる抗体を典型的に含むような、および/または同一なエピトープに対応する異なる抗体を典型的に含むような、通常のポリクローナル抗体とは異なる。その特異性に加えて、モノクローナル抗体は、他の免疫グロブリンによって汚染されていないハイブリドーマ培養から合成できる点で有用である。「モノクローナル」という形容は、実質的に均一な抗体集団から得られるという特徴を示していてもよいが、抗体を何か特定の方法で生産しなければならないことを意味するものではない。例えば、モノクローナル抗体は、"Kohler G, Milstein C.,Nature. 1975 Aug 7;256(5517):495-497."に掲載されているようなハイブリドーマ法と同様の方法によって作製してもよい。あるいは、モノクローナル抗体は、米国特許第4816567号に記載されているような組換え法と同様の方法によって作製してもよい。または、モノクローナル抗体は、"Clackson et al., Nature. 1991 Aug 15;352(6336):624-628."、または"Marks et al., J Mol Biol. 1991 Dec 5;222(3):581-597."に記載されているような技術と同様の方法を用いてファージ抗体ライブラリーから単離してもよい。または、"タンパク質実験ハンドブック,羊土社(2003):92-96."に掲載されている方法によって作製してもよい。
In one embodiment of the present disclosure, a "monoclonal antibody" includes a population in which the individual antibodies are substantially identical to a single epitope, except for antibodies with a small amount of naturally occurring mutations. Alternatively, the individual antibodies in the population may be substantially identical to a single epitope, except for antibodies with a small amount of naturally occurring mutations. Monoclonal antibodies are highly specific and differ from conventional polyclonal antibodies, which typically contain different antibodies that are directed to different epitopes and/or different antibodies that are directed to the same epitope. In addition to their specificity, monoclonal antibodies are useful in that they can be synthesized from hybridoma cultures that are uncontaminated by other immunoglobulins. The term "monoclonal" may indicate the characteristic of being obtained from a substantially homogeneous population of antibodies, but does not imply that the antibody must be produced by any particular method. For example, monoclonal antibodies may be produced by methods similar to the hybridoma method described in "Kohler G, Milstein C., Nature. 1975 Aug 7; 256(5517): 495-497." Alternatively, monoclonal antibodies may be produced by methods similar to the recombinant method described in U.S. Pat. No. 4,816,567. Alternatively, monoclonal antibodies may be isolated from phage antibody libraries using methods similar to the techniques described in "Clackson et al., Nature. 1991 Aug 15; 352(6336): 624-628." or "Marks et al., J Mol Biol. 1991 Dec 5; 222(3): 581-597." Alternatively, monoclonal antibodies may be produced by methods described in "Protein Experiment Handbook, Yodosha (2003): 92-96."
本開示の一実施形態において「キメラ抗体」は、例えば、異種生物間における抗体の可変領域と、抗体の定常領域とを連結したもので、遺伝子組換え技術によって構築できる。マウス-ヒトキメラ抗体は、例えば、"Roguska et al., Proc Natl Acad Sci USA. 1994Feb 1;91(3):969-973."に記載の方法で作製できる。マウス-ヒトキメラ抗体を作製するための基本的な方法は、例えば、クローン化されたcDNAに存在するマウスリーダー配列および可変領域配列を、哺乳類細胞の発現ベクター中にすでに存在するヒト抗体定常領域をコードする配列に連結する。または、クローン化されたcDNAに存在するマウスリーダー配列および可変領域配列をヒト抗体定常領域をコードする配列に連結した後、哺乳類細胞発現ベクターに連結してもよい。ヒト抗体定常領域の断片は、任意のヒト抗体のH鎖定常領域およびヒト抗体のL鎖定常領域のものとすることができ、例えばヒトH鎖のものについてはCγ1、Cγ2、Cγ3またはCγ4を、L鎖のものについてはCλまたはCκを各々挙げることができる。
In one embodiment of the present disclosure, a "chimeric antibody" is, for example, a combination of an antibody variable region and an antibody constant region between different organisms, and can be constructed by recombinant gene technology. A mouse-human chimeric antibody can be produced, for example, by the method described in "Roguska et al., Proc Natl Acad Sci USA. 1994Feb 1;91(3):969-973." The basic method for producing a mouse-human chimeric antibody is, for example, to link a mouse leader sequence and a variable region sequence present in a cloned cDNA to a sequence encoding a human antibody constant region already present in an expression vector for mammalian cells. Alternatively, the mouse leader sequence and the variable region sequence present in the cloned cDNA may be linked to a sequence encoding a human antibody constant region, and then linked to a mammalian cell expression vector. The fragment of the human antibody constant region can be any human antibody H chain constant region or human antibody L chain constant region, for example, Cγ1, Cγ2, Cγ3, or Cγ4 for the human H chain, and Cλ or Cκ for the L chain.
本開示の一実施形態において「ヒト化抗体」は、例えば、非ヒト種由来の1つ以上のCDR、およびヒト免疫グロブリン由来のフレームワーク領域(FR)、さらにヒト免疫グロブリン由来の定常領域を有し、所望の抗原に結合する抗体である。抗体のヒト化は、当該技術分野で既知の種々の手法を使用して実施可能である(Almagro et al., Front Biosci. 2008 Jan 1;13:1619-1633.)。例えば、CDRグラフティング(Ozaki et al., Blood.1999 Jun1;93(11):3922-3930.)、Re-surfacing (Roguska et al., Proc Natl Acad Sci USA.1994 Feb 1;91(3):969-973.)、またはFRシャッフル(Damschroder et al., Mol Immunol. 2007Apr;44(11):3049-3060. Epub 2007 Jan 22.)などが挙げられる。抗原結合を改変するために(好ましくは改善するために)、ヒトFR領域のアミノ酸残基は、CDRドナー抗体からの対応する残基と置換してもよい。このFR置換は、当該技術分野で周知の方法によって実施可能である(Riechmann et al., Nature. 1988 Mar 24;332(6162):323-327.)。例えば、CDRとFR残基の相互作用のモデリングによって抗原結合に重要なFR残基を同定してもよい。または、配列比較によって、特定の位置で異常なFR残基を同定してもよい。
In one embodiment of the present disclosure, a "humanized antibody" is an antibody that has, for example, one or more CDRs from a non-human species, a framework region (FR) from a human immunoglobulin, and a constant region from a human immunoglobulin, and that binds to a desired antigen. Antibody humanization can be performed using various techniques known in the art (Almagro et al., Front Biosci. 2008 Jan 1;13:1619-1633.). For example, CDR grafting (Ozaki et al., Blood. 1999 Jun 1; 93(11): 3922-3930.), Re-surfacing (Roguska et al., Proc Natl Acad Sci USA. 1994 Feb 1; 91(3): 969-973.), or FR shuffling (Damschroder et al., Mol Immunol. 2007 Apr; 44(11): 3049-3060. Epub 2007 Jan 22.) can be used. To alter (preferably improve) antigen binding, amino acid residues in the human FR regions may be replaced with corresponding residues from the CDR donor antibody. This FR substitution can be performed by methods well known in the art (Riechmann et al., Nature. 1988 Mar 24;332(6162):323-327.). For example, FR residues important for antigen binding can be identified by modeling the interactions of CDR and FR residues. Alternatively, unusual FR residues at specific positions can be identified by sequence comparison.
本開示の一実施形態において「ヒト抗体」は、例えば、抗体を構成する重鎖の可変領域および定常領域、軽鎖の可変領域および定常領域を含む領域が、ヒトイムノグロブリンをコードする遺伝子に由来する抗体である。主な作製方法としてはヒト抗体作製用トランスジェニックマウス法、ファージディスプレイ法などがある。ヒト抗体作製用トランスジェニックマウス法では、内因性Igをノックアウトしたマウスに機能的なヒトのIg遺伝子を導入すれば、マウス抗体の代わりに多様な抗原結合能を持つヒト抗体が産生される。さらにこのマウスを免疫すればヒトモノクローナル抗体を従来のハイブリドーマ法で得ることが可能である。例えば、"Lonberg et al., Int Rev Immunol.1995;13(1):65-93."に記載の方法で作製できる。ファージディスプレイ法は、典型的には大腸菌ウイルスの一つであるM13やT7などの繊維状ファージのコート蛋白質(g3p、g10p等)のN末端側にファージの感染性を失わないよう外来遺伝子を融合蛋白質として発現させるシステムである。例えば、"Vaughan et al., Nat Biotechnol. 1996 Mar;14(3):309-314."に記載の方法で作製できる。
In one embodiment of the present disclosure, a "human antibody" is, for example, an antibody in which the regions constituting the antibody, including the variable and constant regions of the heavy chain and the variable and constant regions of the light chain, are derived from genes encoding human immunoglobulin. Major production methods include the transgenic mouse method for producing human antibodies and the phage display method. In the transgenic mouse method for producing human antibodies, if a functional human Ig gene is introduced into a mouse in which endogenous Ig has been knocked out, human antibodies with diverse antigen-binding abilities are produced instead of mouse antibodies. Furthermore, if this mouse is immunized, human monoclonal antibodies can be obtained by the conventional hybridoma method. For example, they can be produced by the method described in "Lonberg et al., Int Rev Immunol. 1995; 13(1): 65-93." The phage display method is a system in which a foreign gene is expressed as a fusion protein on the N-terminus of the coat protein (g3p, g10p, etc.) of a filamentous phage, typically an E. coli virus such as M13 or T7, so that the phage does not lose its infectivity. For example, it can be produced by the method described in "Vaughan et al., Nat Biotechnol. 1996 Mar;14(3):309-314."
本明細書において、抗体の「抗原結合断片」とは、抗体が抗原に結合する機能を維持した任意の長さの断片を指す。抗原結合断片としては、例えば、Fab、F(ab’)2、Fab’、Fv、scFvなどが挙げられるが、これらに限定されない。
As used herein, the term "antigen-binding fragment" of an antibody refers to a fragment of any length that maintains the antibody's ability to bind to an antigen. Examples of antigen-binding fragments include, but are not limited to, Fab, F(ab') 2 , Fab', Fv, and scFv.
本明細書において、「融合タンパク質」とは、2種類以上の異なるタンパク質またはその断片が共有結合した、またはそれらの遺伝子が一体となって組換え発現された1個のタンパク質を指す。
As used herein, the term "fusion protein" refers to a single protein in which two or more different proteins or fragments thereof are covalently linked, or in which the genes of these proteins are recombinantly expressed together.
本明細書において、「被験体由来の細胞」とは、本開示の組成物、医薬、細胞製剤もしくは細胞が投与される、または本開示の方法の対象となる被験体から得た細胞または該被験体から得た細胞に由来する細胞をいう。本明細書において、「被験体由来の抗原」とは、免疫応答を生じさせる被験体自身が生成する抗原、例えば、自己免疫疾患を有する被験体における自己免疫疾患の原因となる被験体自身が生成する抗原をいう。本明細書において、「被験体に由来しない抗原」とは、免疫応答を生じさせ得る外来の抗原をいう。
As used herein, "cells derived from a subject" refers to cells obtained from a subject to which the composition, pharmaceutical, cell preparation, or cells disclosed herein is administered or which is the subject of the method disclosed herein, or cells derived from cells obtained from the subject. As used herein, "antigen derived from a subject" refers to an antigen produced by the subject itself that elicits an immune response, for example, an antigen produced by the subject itself that causes an autoimmune disease in a subject with an autoimmune disease. As used herein, "antigen not derived from a subject" refers to a foreign antigen that can elicit an immune response.
本明細書において、「被験体に由来しない抗原の含有物」は、被験体に由来しない抗原を含有する任意の物質または物質の集合物をいい、例えば、被験体に由来しない抗原を発現している細胞、細胞集団、組織等が挙げられる。
As used herein, "a substance containing an antigen not derived from a subject" refers to any substance or collection of substances that contains an antigen not derived from a subject, such as a cell, cell population, tissue, etc. that expresses an antigen not derived from a subject.
本明細書において、「移植免疫拒絶反応」とは、臓器、組織または細胞の移植を受けた被験体において、被験体の免疫系が、移植された臓器、組織または細胞に対して攻撃し、損傷または破壊することをいう。
As used herein, "transplant immune rejection" refers to a condition in which the immune system of a subject who has received an organ, tissue, or cell transplant attacks and damages or destroys the transplanted organ, tissue, or cell.
本明細書において、「アレルギー」とは、被験体に由来しない抗原に対して、免疫応答が過剰に起こる状態をいう。アレルギーを引き起こす被験体に由来しない抗原は、アレルゲンとも呼ばれ、例えば、ダニ抗原、卵白抗原、ミルク抗原、小麦抗原、ピーナッツ抗原、大豆抗原、ソバ抗原、ゴマ抗原、コメ抗原、甲殻類抗原、キウイ抗原、リンゴ抗原、バナナ抗原、モモ抗原、トマト抗原、マグロ抗原、サケ抗原、サバ抗原、牛肉抗原、鶏肉抗原、豚肉抗原、ネコ皮屑抗原、昆虫抗原、花粉抗原、イヌ皮屑抗原、真菌抗原、細菌抗原、ラテックス、ハプテンおよび金属等が挙げられるがこれらに限定されない。
In this specification, "allergy" refers to a state in which an excessive immune response occurs against an antigen not derived from the subject. Antigens not derived from the subject that cause allergies are also called allergens, and examples of such antigens include, but are not limited to, dust mite antigens, egg white antigens, milk antigens, wheat antigens, peanut antigens, soybean antigens, buckwheat antigens, sesame antigens, rice antigens, crustacean antigens, kiwi antigens, apple antigens, banana antigens, peach antigens, tomato antigens, tuna antigens, salmon antigens, mackerel antigens, beef antigens, chicken antigens, pork antigens, cat dander antigens, insect antigens, pollen antigens, dog dander antigens, fungal antigens, bacterial antigens, latex, haptens, and metals.
本明細書において、「自己免疫疾患」とは、免疫系が自身の細胞、組織または臓器に対して望ましくない免疫応答を行う任意の疾患をいう。自己免疫疾患としては、例えば、関節リウマチ、多発性硬化症、1型糖尿病、炎症性腸疾患(例えば、クローン病または潰瘍性大腸炎)、全身性エリテマトーデス、乾癬、強皮症、自己免疫性甲状腺疾患、円形脱毛症、グレーブス病、ギラン・バレー症候群、セリアック病、シェーグレン症候群、リウマチ熱、胃炎、自己免疫性萎縮性胃炎、自己免疫性肝炎、膵島炎、卵巣炎、精巣炎、ブドウ膜炎、水晶体起因性ブドウ膜炎、重症筋無力症、原発性粘液水腫、悪性貧血、自己免疫性溶血性貧血、アジソン病、強皮症、グッドパスチャー症候群、腎炎(例えば、糸球体腎炎)、乾癬、尋常性天疱瘡、類天疱瘡、交感性眼炎、特発性血小板減少性紫斑病、特発性白血球減少症、ウェゲナー肉芽腫および多発性/皮膚筋炎が挙げられるが、これらに限定されない。
As used herein, "autoimmune disease" refers to any disease in which the immune system mounts an unwanted immune response against one's own cells, tissues, or organs. Examples of autoimmune diseases include rheumatoid arthritis, multiple sclerosis, type 1 diabetes, inflammatory bowel disease (e.g., Crohn's disease or ulcerative colitis), systemic lupus erythematosus, psoriasis, scleroderma, autoimmune thyroid disease, alopecia areata, Graves' disease, Guillain-Barré syndrome, celiac disease, Sjögren's syndrome, rheumatic fever, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, insulitis, oophoritis, and sepsis. These include, but are not limited to, focal ulcers, uveitis, phacogenic uveitis, myasthenia gravis, primary myxedema, pernicious anemia, autoimmune hemolytic anemia, Addison's disease, scleroderma, Goodpasture's syndrome, nephritis (e.g., glomerulonephritis), psoriasis, pemphigus vulgaris, pemphigoid, sympathetic ophthalmia, idiopathic thrombocytopenic purpura, idiopathic leukopenia, Wegener's granulomatosis, and poly/dermatomyositis.
本明細書において、「移植片対宿主病」とは、移植された臓器、組織または細胞が、免疫応答によって、移植を受けた被験体の細胞、組織または臓器を攻撃し、損傷または破壊することをいう。
As used herein, "graft-versus-host disease" refers to an immune response in which a transplanted organ, tissue, or cell attacks, damages, or destroys the cells, tissues, or organs of the recipient.
本明細書において、「iPS細胞もしくはES細胞またはそれらの細胞から分化した細胞、組織もしくは臓器の移植によって引き起こされる免疫拒絶反応」とは、iPS細胞もしくはES細胞が有する抗原、またはiPS細胞もしくはES細胞から分化した細胞、組織もしくは臓器が有する抗原によって生じる免疫拒絶反応をいう。
In this specification, "immune rejection caused by transplantation of iPS cells or ES cells, or cells, tissues, or organs differentiated from these cells" refers to an immune rejection caused by an antigen present in iPS cells or ES cells, or an antigen present in cells, tissues, or organs differentiated from iPS cells or ES cells.
本明細書において、「制御性T細胞(regulatory T cell)(Tregとも称される)」とは、抗原に対する免疫応答を制御するT細胞を指す。制御性T細胞は、Foxp3陽性であり得る。制御性T細胞はCD4陽性であっても、CD8陽性であってもよい。典型的には、制御性T細胞は、Foxp3陽性、CD25陽性およびCD4陽性であり得る。
As used herein, "regulatory T cells (also referred to as Tregs)" refer to T cells that control the immune response to an antigen. Regulatory T cells may be Foxp3 positive. Regulatory T cells may be CD4 positive or CD8 positive. Typically, regulatory T cells may be Foxp3 positive, CD25 positive, and CD4 positive.
本明細書において、「抑制性T細胞(suppressor T cell)」とは、抗原に対する免疫応答を抑制する、および/または抗原に対する免疫応答を示さないもしくは抑制されたT細胞を指す。
As used herein, "suppressor T cell" refers to a T cell that suppresses the immune response to an antigen and/or that does not show or has a suppressed immune response to an antigen.
本明細書において、「抗原特異的抑制性T細胞」とは、特定の抗原に対する免疫応答を特異的に抑制する、および/または特定の抗原に対する特異的免疫反応を示さないもしくは抑制されたT細胞を指す。
As used herein, "antigen-specific suppressor T cells" refers to T cells that specifically suppress immune responses to a specific antigen and/or T cells that do not show or are suppressed from showing a specific immune response to a specific antigen.
本明細書において、「誘導型抑制性T細胞(induced suppressor T cell)」とは、本開示の方法、組成物、または因子による刺激によって誘導される抑制性T細胞であり、制御性T細胞(例えばFOXP3陽性CD4陽性CD25陽性T細胞)および抑制性T細胞(例えば、CD4陽性アナジーT細胞またはCD8陽性アナジーT細胞)を含む細胞集団を指す。CD8陽性アナジー細胞およびCD4陽性アナジー細胞には、CD44陽性の細胞を多く含み、CD8陽性細胞および/またはCD4陽性細胞は、CD44陽性であり得る。また、アナジー細胞の生成の確認には、CD44の他に、CD45RA/CD45ROも使用することができる。例えば、CD8陽性アナジーT細胞および/またはCD4陽性アナジーT細胞はCD45RA陰性かつCD45RO陽性である。
As used herein, "induced suppressor T cells" refers to suppressor T cells induced by stimulation with the methods, compositions, or factors disclosed herein, and refers to a cell population including regulatory T cells (e.g., FOXP3-positive CD4-positive CD25-positive T cells) and suppressor T cells (e.g., CD4-positive anergy T cells or CD8-positive anergy T cells). CD8-positive anergy cells and CD4-positive anergy cells contain many CD44-positive cells, and CD8-positive cells and/or CD4-positive cells may be CD44-positive. In addition to CD44, CD45RA/CD45RO can also be used to confirm the generation of anergy cells. For example, CD8-positive anergy T cells and/or CD4-positive anergy T cells are CD45RA-negative and CD45RO-positive.
本明細書において、「抗原特異的誘導型抑制性T細胞」とは、特定の抗原に対する免疫応答を特異的に抑制する、および/または特定の抗原に対する特異的免疫反応を示さないもしくは抑制された誘導型抑制性T細胞を指す。
As used herein, "antigen-specific induced suppressor T cells" refers to induced suppressor T cells that specifically suppress immune responses to a specific antigen and/or that do not show or are suppressed to show a specific immune response to a specific antigen.
本明細書において、「抗原特異的誘導型抑制性T細胞を選択的に誘導」とは、必ずしも誘導型抑制性T細胞が100%抗原特異的である必要はなく、本開示の方法、組成物または因子による刺激をしない場合(例えば、抗原特異的誘導型抑制性T細胞を選択的に誘導する因子と細胞とを接触させない場合)と比べて、本開示の方法、組成物または因子による刺激をした場合(例えば、抗原特異的誘導型抑制性T細胞を選択的に誘導する因子と細胞とを接触させる場合)、抗原特異的誘導型抑制性T細胞の割合が増加するように誘導することを指す。
As used herein, "selectively inducing antigen-specific induced suppressive T cells" does not necessarily require that the induced suppressive T cells be 100% antigen-specific, but refers to inducing an increase in the proportion of antigen-specific induced suppressive T cells when stimulated with the method, composition, or factor disclosed herein (for example, when cells are contacted with a factor that selectively induces antigen-specific induced suppressive T cells) compared to when not stimulated with the method, composition, or factor disclosed herein (for example, when cells are not contacted with a factor that selectively induces antigen-specific induced suppressive T cells).
本明細書において、「抗原特異的誘導型抑制性T細胞を選択的に誘導する因子」とは、必ずしも誘導型抑制性T細胞が100%抗原特異的である必要はなく、この因子を細胞と接触させない場合と比べて、抗原特異的誘導型抑制性T細胞の割合が増加するように誘導する任意の因子を指す。
In this specification, the term "factor that selectively induces antigen-specific induced suppressor T cells" refers to any factor that induces an increase in the proportion of antigen-specific induced suppressor T cells compared to when the factor is not in contact with cells, and does not necessarily require that the induced suppressor T cells be 100% antigen-specific.
(好ましい実施形態)
以下に好ましい実施形態の説明を記載するが、この実施形態は本開示の例示であり、本開示の範囲はそのような好ましい実施形態に限定されないことが理解されるべきである。当業者はまた、以下のような好ましい実施例を参考にして、本開示の範囲内にある改変、変更などを容易に行うことができることが理解されるべきである。これらの実施形態について、当業者は適宜、本明細書中の記載を参酌して、任意の実施形態を組み合わせ得る。また、本開示の以下の実施形態は単独でも使用されあるいはそれらを組み合わせて使用することができることが理解される。 Preferred Embodiments
A description of a preferred embodiment is given below, but it should be understood that this embodiment is an example of the present disclosure and the scope of the present disclosure is not limited to such a preferred embodiment. It should also be understood that those skilled in the art can easily make modifications, changes, etc. within the scope of the present disclosure by referring to the following preferred examples. Regarding these embodiments, those skilled in the art can combine any embodiment as appropriate, taking into consideration the description in this specification. It is also understood that the following embodiments of the present disclosure can be used alone or in combination.
以下に好ましい実施形態の説明を記載するが、この実施形態は本開示の例示であり、本開示の範囲はそのような好ましい実施形態に限定されないことが理解されるべきである。当業者はまた、以下のような好ましい実施例を参考にして、本開示の範囲内にある改変、変更などを容易に行うことができることが理解されるべきである。これらの実施形態について、当業者は適宜、本明細書中の記載を参酌して、任意の実施形態を組み合わせ得る。また、本開示の以下の実施形態は単独でも使用されあるいはそれらを組み合わせて使用することができることが理解される。 Preferred Embodiments
A description of a preferred embodiment is given below, but it should be understood that this embodiment is an example of the present disclosure and the scope of the present disclosure is not limited to such a preferred embodiment. It should also be understood that those skilled in the art can easily make modifications, changes, etc. within the scope of the present disclosure by referring to the following preferred examples. Regarding these embodiments, those skilled in the art can combine any embodiment as appropriate, taking into consideration the description in this specification. It is also understood that the following embodiments of the present disclosure can be used alone or in combination.
本発明者らは、誘導型抑制性T細胞の誘導において、増殖因子であるIL-2の産生または産生されたIL-2の機能を調節することによって、誘導型抑制性T細胞に抗原特異的抑制能が誘導されることを見出した。さらに、本発明者らは、誘導型抑制性T細胞の抗原非特異的な分裂増殖を抑える目的でIL-2産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子を添加し、抗原特異性を誘導する新しい培養方法も見出した。
The inventors have found that in the induction of inducible suppressor T cells, regulating the production of the growth factor IL-2 or the function of the produced IL-2 induces antigen-specific suppressive ability in the inducible suppressor T cells. Furthermore, the inventors have also found a new culture method for inducing antigen specificity by adding a regulatory factor capable of regulating IL-2 production or a regulatory factor capable of regulating the function of the produced IL-2, with the aim of suppressing the antigen-nonspecific division and proliferation of inducible suppressor T cells.
一態様において、本開示は、被験体由来の細胞から抗原特異的誘導型抑制性T細胞を選択的に誘導させる方法であって、抗原特異的誘導型抑制性T細胞を選択的に誘導する因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合する工程を含む、方法を提供する。抗原の含有物は細胞であり得、該細胞の増殖および活性化を防止するために、放射線照射され得る。
In one aspect, the present disclosure provides a method for selectively inducing antigen-specific induced suppressor T cells from cells derived from a subject, the method comprising the step of mixing a factor that selectively induces antigen-specific induced suppressor T cells, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen. The substance containing the antigen may be a cell, which may be irradiated to prevent proliferation and activation of the cell.
別の態様において、本開示は、被験体由来の細胞から誘導型抑制性T細胞を作製する方法であって、IL-2の産生を阻害することができる阻害因子または産生されたIL-2の機能を阻害することができる阻害因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合する工程を含む、方法を提供する。
In another aspect, the present disclosure provides a method for producing induced suppressor T cells from cells derived from a subject, the method comprising the step of mixing an inhibitor capable of inhibiting the production of IL-2 or an inhibitor capable of inhibiting the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
別の態様において、本開示は、被験体由来の細胞からの誘導型抑制性T細胞の作製において、特定の抗原に対して非特異的な誘導型抑制性T細胞の増殖を抑制するための方法であって、抗原特異的誘導型抑制性T細胞を選択的に誘導する因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合する工程を含む、方法を提供する。
In another aspect, the present disclosure provides a method for suppressing the proliferation of induced suppressive T cells non-specific to a specific antigen in the production of induced suppressive T cells from cells derived from a subject, the method comprising the step of mixing a factor that selectively induces antigen-specific induced suppressive T cells, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
いくつかの実施形態において、本開示の方法は、上記因子、被験体由来の細胞、および被験体由来の抗原あるいは被験体に由来しない抗原または抗原の含有物を含む混合物を培養する工程を含む。培養後、本開示の誘導型抑制性T細胞は、CD4陽性アナジーT細胞、CD8陽性T細胞、および制御性T細胞を含み得る。制御性T細胞は、Foxp3+などの細胞表面マーカーを示し、好ましくはCD4+、CD25+、及びFoxp3+などの細胞表面マーカーを示す。
In some embodiments, the method of the present disclosure includes culturing a mixture containing the above-mentioned factor, cells derived from a subject, and antigens derived from the subject or antigens not derived from the subject or antigens contained therein. After culturing, the induced suppressor T cells of the present disclosure can include CD4 positive anergy T cells, CD8 positive T cells, and regulatory T cells. The regulatory T cells exhibit cell surface markers such as Foxp3 + , preferably CD4 + , CD25 + , and Foxp3 + .
いくつかの実施形態において、本開示の方法、因子および組成物を用いた培養の培養終了時の培養物中、CD4+T細胞の割合は、全細胞に対して約20%以上、約25%以上、約30%以上、約35%以上、約40%以上、約45%以上、または約50%以上であり得る。好ましい実施形態において、培養物中、CD4+T細胞の割合は、全細胞に対して約35%以上であり得る。
In some embodiments, the percentage of CD4 + T cells in the culture at the end of the culture using the disclosed methods, factors, and compositions can be about 20% or more, about 25% or more, about 30% or more, about 35% or more, about 40% or more, about 45% or more, or about 50% or more of the total cells. In a preferred embodiment, the percentage of CD4 + T cells in the culture can be about 35% or more of the total cells.
さらなる実施形態において、本開示の方法、因子および組成物を用いた培養の培養終了時の培養物中、制御性T細胞(例えば、CD4+Foxp3+T細胞)の割合は、全細胞に対して約5%以上、約10%以上、約15%以上、約20%以上、または約25%以上であり得る。好ましい実施形態において、培養物中、制御性T細胞の割合は、全細胞に対して約15%以上であり得る。
In further embodiments, the percentage of regulatory T cells (e.g., CD4 + Foxp3 + T cells) in the culture at the end of culture using the disclosed methods, factors, and compositions may be about 5% or more, about 10% or more, about 15% or more, about 20% or more, or about 25% or more of total cells. In a preferred embodiment, the percentage of regulatory T cells in the culture may be about 15% or more of total cells.
いくつかの実施形態において、本開示の方法、因子および組成物により、CD4+T細胞中のCD4+制御性T細胞(例えば、CD4+Foxp3+T細胞)の割合が増加し得る。本開示の方法、因子および組成物を用いた培養の培養終了時の培養物において、CD4+T細胞中のCD4+制御性T細胞(例えば、CD4+Foxp3+T細胞)の割合は、約30%以上、約35%以上、約40%以上、約45%以上、約50%以上、約55%以上、約60%以上、約65%以上、約70%以上、約80%以上、約90%以上、約100%であり得る。好ましい実施形態において、本開示の方法、因子および組成物を用いた培養の培養終了時の培養物において、CD4+T細胞中のCD4+制御性T細胞(例えば、CD4+Foxp3+T細胞)の割合は、約50%以上であり得る。
In some embodiments, the disclosed methods, factors, and compositions can increase the proportion of CD4 + regulatory T cells (e.g., CD4 + Foxp3 + T cells) among CD4 + T cells. In the culture at the end of culture using the disclosed methods, factors, and compositions, the proportion of CD4 + regulatory T cells (e.g., CD4 + Foxp3 + T cells) among CD4 + T cells can be about 30% or more, about 35% or more, about 40% or more, about 45% or more, about 50% or more, about 55% or more, about 60% or more, about 65% or more, about 70% or more, about 80% or more, about 90% or more, or about 100%. In a preferred embodiment, the percentage of CD4 + regulatory T cells (e.g., CD4 + Foxp3 + T cells) among CD4 + T cells in cultures at the end of culture using the methods, factors, and compositions of the present disclosure may be about 50% or greater.
いくつかの実施形態において、前記細胞は、末梢血単核球(PBMC)、脾臓細胞、骨髄細胞、リンパ節細胞、またこれらの任意の組み合わせであり得る。
In some embodiments, the cells can be peripheral blood mononuclear cells (PBMCs), spleen cells, bone marrow cells, lymph node cells, or any combination thereof.
いくつかの実施形態において、本開示の方法は、インターロイキン-2(IL-2)を添加することなく実施される。
In some embodiments, the methods of the present disclosure are performed without the addition of interleukin-2 (IL-2).
いくつかの実施形態において、抗原特異的誘導型抑制性T細胞を選択的に誘導する因子は、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子であり得る。前記調節は、好ましくは負の調節であり得る。特定の理論に束縛されることは望まないが、IL-2は、特定の抗原に対して非特異的な誘導型抑制性T細胞の増殖も促進するため、IL-2の産生を調節するか、産生されたIL-2の機能を調節することによって、非特異的な誘導型抑制性T細胞の増殖を抑制することが可能である。IL-2の産生および機能を完全に阻害すると抗原特異的誘導型抑制性T細胞の増殖も抑制してしまうため、所定量のIL-2を存在することが好ましい。
In some embodiments, the factor that selectively induces antigen-specific induced suppressor T cells may be a regulator capable of regulating the production of IL-2 or a regulator capable of regulating the function of the produced IL-2. The regulation may preferably be a negative regulation. Without wishing to be bound by a particular theory, IL-2 also promotes the proliferation of induced suppressor T cells that are non-specific to a particular antigen, so it is possible to suppress the proliferation of non-specific induced suppressor T cells by regulating the production of IL-2 or regulating the function of the produced IL-2. Since complete inhibition of the production and function of IL-2 would also suppress the proliferation of antigen-specific induced suppressor T cells, it is preferable that a certain amount of IL-2 is present.
いくつかの実施形態において、所定量のIL-2は、培養物中、約100pg/mL以下、約90pg/mL以下、約80pg/mL以下、約70pg/mL以下、約60pg/mL以下、約50pg/mL以下、約40pg/mL以下、約30pg/mL以下、約20pg/mL以下、約19pg/mL以下、約18pg/mL以下、約17pg/mL以下、約16pg/mL以下、約15pg/mL以下、約14pg/mL以下、約13pg/mL以下、約12pg/mL以下、約11pg/mL以下、約10pg/mL以下、約9pg/mL以下、約8pg/mL以下、約7pg/mL以下、約6pg/mL以下、または約5pg/mL以下であり得る。上記所定量は、約0.5~2×106個/mLの細胞濃度でのIL-2の量であり、IL-2の量は、培養中の細胞数に応じて適宜変更し得る。所定量のIL-2は、起源によっても異なり得る。適切な所定量のIL-2は、実施例3および6に基づき、適宜決定することができる。
In some embodiments, the predetermined amount of IL-2 can be about 100 pg/mL or less, about 90 pg/mL or less, about 80 pg/mL or less, about 70 pg/mL or less, about 60 pg/mL or less, about 50 pg/mL or less, about 40 pg/mL or less, about 30 pg/mL or less, about 20 pg/mL or less, about 19 pg/mL or less, about 18 pg/mL or less, about 17 pg/mL or less, about 16 pg/mL or less, about 15 pg/mL or less, about 14 pg/mL or less, about 13 pg/mL or less, about 12 pg/mL or less, about 11 pg/mL or less, about 10 pg/mL or less, about 9 pg/mL or less, about 8 pg/mL or less, about 7 pg/mL or less, about 6 pg/mL or less, or about 5 pg/mL or less in culture. The above-mentioned predetermined amount is the amount of IL-2 at a cell concentration of about 0.5 to 2×10 6 cells/mL, and the amount of IL-2 can be appropriately changed depending on the number of cells being cultured. The predetermined amount of IL-2 can also differ depending on the source. An appropriate predetermined amount of IL-2 can be appropriately determined based on Examples 3 and 6.
いくつかの実施形態において、培養中のIL-2の所定量を達成するために、培養の開始時または途中で、適宜IL-2を添加してもよく、あるいは抗IL-2抗体を添加してもよい。
In some embodiments, IL-2 or an anti-IL-2 antibody may be added at the beginning or during the culture to achieve a predetermined amount of IL-2 during the culture.
いくつかの実施形態において、本開示の方法または因子によって製造された抗原特異的誘導型抑制性T細胞またはこれを含む細胞集団の抗原に対する抑制能は、本開示の方法または因子を使用せずに培養したT細胞またはその細胞集団による免疫反応を基準として、約30%以上(すなわち、基準に対して70%以下の免疫反応)、約40%以上、約50%以上、約60%以上、約70%以上、約80%以上、約90%以上、約95%以上、約96%以上、約97%以上、約98%以上、約99%以上であり得る、好ましい実施形態において、本開示の方法または因子によって製造された抗原特異的誘導型抑制性T細胞またはこれを含む細胞集団の抗原に対する抑制能は、約50%以上であり得る。
In some embodiments, the suppressive ability of antigen-specific induced suppressive T cells or cell populations containing the same produced by the method or factor of the present disclosure against an antigen may be about 30% or more (i.e., an immune response of 70% or less relative to the standard), about 40% or more, about 50% or more, about 60% or more, about 70% or more, about 80% or more, about 90% or more, about 95% or more, about 96% or more, about 97% or more, about 98% or more, or about 99% or more, based on the immune response of T cells or cell populations cultured without using the method or factor of the present disclosure. In a preferred embodiment, the suppressive ability of antigen-specific induced suppressive T cells or cell populations containing the same produced by the method or factor of the present disclosure against an antigen may be about 50% or more.
いくつかの実施形態において、抗原特異的誘導型抑制性T細胞を選択的に誘導する因子は、IL-2の産生を調節もしくは阻害することができる調節因子もしくは阻害因子または産生されたIL-2の機能を調節もしくは阻害することができる調節因子または阻害因子、またはその両方の機能を有する因子であり得る。
In some embodiments, the factor that selectively induces antigen-specific induced suppressor T cells may be a regulator or inhibitor that can regulate or inhibit the production of IL-2, or a regulator or inhibitor that can regulate or inhibit the function of the produced IL-2, or a factor that has both functions.
いくつかの実施形態において、抗原特異的誘導型抑制性T細胞を選択的に誘導する因子は、IL-2とIL-2受容体(IL-2R)との相互作用を阻害する阻害因子、ならびに/あるいは、CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子であり得る。
In some embodiments, the factor that selectively induces antigen-specific induced suppressor T cells can be an inhibitory factor that inhibits the interaction between IL-2 and the IL-2 receptor (IL-2R) and/or an inhibitory factor that can inhibit the interaction between CD80 and/or CD86 and CD28.
IL-2とIL-2受容体(IL-2R)との相互作用を阻害することができる阻害因子としては、例えば、抗IL-2抗体、抗IL-2R抗体、抗IL-2Rα(CD25)抗体、抗IL-2Rβ(CD122)抗体またはこれらの抗原結合断片などが挙げられるが、これらに限定されない。
Examples of inhibitors capable of inhibiting the interaction between IL-2 and IL-2 receptor (IL-2R) include, but are not limited to, anti-IL-2 antibodies, anti-IL-2R antibodies, anti-IL-2Rα (CD25) antibodies, anti-IL-2Rβ (CD122) antibodies, or antigen-binding fragments thereof.
IL-2の産生を阻害することができる阻害因子としては、CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子、またはIL-2産生を阻害する小分子化合物などが挙げられる。
Examples of inhibitors that can inhibit IL-2 production include inhibitors that can inhibit the interaction between CD80 and/or CD86 and CD28, or small molecule compounds that inhibit IL-2 production.
産生されたIL-2の機能を調節もしくは阻害することができる調節因子もしくは阻害因子としては、IL-2とIL-2受容体(IL-2R)との相互作用を阻害することができる阻害因子、IL-2の作用を阻害する小分子化合物などが挙げられるが、これらに限定されない。
Regulatory or inhibitory factors capable of regulating or inhibiting the function of the produced IL-2 include, but are not limited to, inhibitory factors capable of inhibiting the interaction between IL-2 and the IL-2 receptor (IL-2R), small molecule compounds that inhibit the action of IL-2, etc.
IL-2産生を阻害する小分子化合物としては、カルシニューリン阻害剤、ステロイド性抗炎症薬などが挙げられるが、これらに限定されない。カルシニューリン阻害剤としては、タクロリムス、シクロスポリンなどが挙げられる。カルシニューリンは、NFAT(活性化T細胞の核因子)を脱リン酸化し、それにより、NFATは、核に侵入してインターロイキン-2促進剤に結合することが可能となる。このプロセスを遮断することにより、IL-2の産生を阻害する。ステロイド性抗炎症薬としては、フランカルボン酸モメタゾン、クロベタゾールプロピオン酸エステル、エタボン酸ロテプレドノール、ジフルプレドナート、デキサメタゾン、アムシノニド、フルランドレノリド、プレドニゾロン、フルオシノロンアセトニド、デソニド、トリアムシノロンアセトニド、ブデソニド、フルドロコルチゾン酢酸エステル、フルオシノニド、メチルプレドニゾロン、ベタメタゾン、デソキシメタゾン、ハルシノニド、フルオロメトロン、ジプロピオン酸ベクロメタゾン、およびデュタステリドなどが挙げられるが、これらに限定されない。IL-2の作用を阻害する小分子化合物としては、ラパマイシン(シロリムス)、SDZ RADなどが挙げられる。ラパマイシンは、細胞内シグナル伝達と細胞増殖を抑制して、IL-2に対するリンパ球の応答を阻害してTリンパ球の活性化を抑止する。
Small molecule compounds that inhibit IL-2 production include, but are not limited to, calcineurin inhibitors, steroidal anti-inflammatory drugs, and the like. Calcineurin inhibitors include tacrolimus, cyclosporine, and the like. Calcineurin dephosphorylates NFAT (nuclear factor of activated T cells), which allows NFAT to enter the nucleus and bind to interleukin-2 promoters. Blocking this process inhibits IL-2 production. Steroidal anti-inflammatory drugs include, but are not limited to, mometasone furoate, clobetasol propionate, loteprednol etabonate, difluprednate, dexamethasone, amcinonide, flurandrenolide, prednisolone, fluocinolone acetonide, desonide, triamcinolone acetonide, budesonide, fludrocortisone acetate, fluocinonide, methylprednisolone, betamethasone, desoximethasone, halcinonide, fluorometholone, beclomethasone dipropionate, and dutasteride. Small molecule compounds that inhibit the action of IL-2 include rapamycin (sirolimus) and SDZ RAD. Rapamycin suppresses intracellular signaling and cell proliferation, inhibiting the response of lymphocytes to IL-2 and suppressing the activation of T lymphocytes.
CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子としては、例えば、抗CD80抗体、抗CD86抗体、CD80およびCD86に対する二重特異性抗体、抗CD28抗体もしくはこれらの抗原結合断片、CTLA4-Ig融合タンパク質、CD28-Ig融合タンパク質などが挙げられるが、これらに限定されない。CTLA4-Ig融合タンパク質は、アバタセプトまたはベラタセプトであり得る。
Inhibitory factors capable of inhibiting the interaction of CD80 and/or CD86 with CD28 include, but are not limited to, for example, anti-CD80 antibodies, anti-CD86 antibodies, bispecific antibodies against CD80 and CD86, anti-CD28 antibodies or antigen-binding fragments thereof, CTLA4-Ig fusion proteins, and CD28-Ig fusion proteins. The CTLA4-Ig fusion protein may be abatacept or belatacept.
いくつかの実施形態において、CD80および/またはCD86は、抗原提示細胞により発現され、CD28は、T細胞により発現される。特定の実施形態において、CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子は、抗CD80抗体および/または抗CD86抗体あるいはCTLA4-Ig融合タンパク質であり得る。本開示における使用が想定される阻害因子としては、上述されるようにCTLA-4 Ig融合タンパク質が挙げられる。CTLA-4 Ig融合タンパク質は、抗原提示細胞上におけるCD80/CD86への結合について、T細胞上の共刺激受容体であるCD28と競合し、その結果、T細胞の活性化を阻害するように機能する。本開示では、上記CTLA-4 Ig融合タンパク質として、アバタセプト(Orencia(登録商標))、ベラタセプトまたはMaxy-4が想定される。ベラタセプトは、CD80およびCD86に対する結合アビディティを顕著に増大させる2つのアミノ酸置換(L104EおよびA29Y)を含有する(Davies JKら、Cell Transplant.(2012);21(9):2047~61、Adams ABら、J Immunol.(2016)197(6):2045~50を参照のこと)。また、CTLA4-Ig融合タンパク質と同様の効果が期待される阻害因子として、CD28-Ig融合タンパク質(Peach RJら、J Exp Med.(1994)180(6):2049~2058)も挙げられる。本開示の阻害因子は、核酸の形態でも使用され得る。一例を挙げると、CTLA4-Ig融合タンパク質をコードする核酸を、アデノウイルスベクターなどを介して細胞に導入し、発現させることも想定される。例えば、Jin YZら、Transplant Proc.(2003);35(8):3156~9を参照のこと。
In some embodiments, CD80 and/or CD86 are expressed by antigen-presenting cells, and CD28 is expressed by T cells. In certain embodiments, the inhibitor capable of inhibiting the interaction of CD80 and/or CD86 with CD28 may be an anti-CD80 antibody and/or an anti-CD86 antibody or a CTLA4-Ig fusion protein. Inhibitors contemplated for use in the present disclosure include CTLA-4 Ig fusion proteins as described above. CTLA-4 Ig fusion proteins function to compete with CD28, a costimulatory receptor on T cells, for binding to CD80/CD86 on antigen-presenting cells, thereby inhibiting T cell activation. In the present disclosure, the CTLA-4 Ig fusion protein is contemplated to be abatacept (Orencia®), belatacept, or Maxy-4. Belatacept contains two amino acid substitutions (L104E and A29Y) that significantly increase the binding avidity for CD80 and CD86 (see Davies JK et al., Cell Transplant. (2012); 21(9): 2047-61; Adams AB et al., J Immunol. (2016) 197(6): 2045-50). In addition, an inhibitor that is expected to have the same effect as the CTLA4-Ig fusion protein is the CD28-Ig fusion protein (Peach RJ et al., J Exp Med. (1994) 180(6): 2049-2058). The inhibitors of the present disclosure may also be used in the form of nucleic acids. For example, it is envisioned that a nucleic acid encoding a CTLA4-Ig fusion protein can be introduced into a cell via an adenovirus vector or the like and expressed. See, for example, Jin YZ et al., Transplant Proc. (2003); 35(8): 3156-9.
さらなる態様において、被験体由来の細胞から抗原特異的誘導型抑制性T細胞を選択的に誘導させるための組成物であって、該組成物は、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子を含み、該組成物が、該被験体由来の細胞、および該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物と接触させられることを特徴とする、組成物を提供する。
In a further aspect, a composition for selectively inducing antigen-specific inducible suppressor T cells from cells derived from a subject is provided, the composition comprising a regulator capable of regulating the production of IL-2 or a regulator capable of regulating the function of the produced IL-2, the composition being contacted with cells derived from the subject and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
(自己由来制御性T細胞製造手順)
以下に、自己由来誘導型抑制性T細胞の典型的な製造方法を説明する。 Autologous Regulatory T Cell Manufacturing Procedure
A typical method for producing autologous induced suppressor T cells is described below.
以下に、自己由来誘導型抑制性T細胞の典型的な製造方法を説明する。 Autologous Regulatory T Cell Manufacturing Procedure
A typical method for producing autologous induced suppressor T cells is described below.
事前の確認
一つの実施形態では、事前の確認は以下のように行うことができる。以下に例示する各種数値、試薬、手順等は、代表例であり、当業者は適宜変更して事前の確認の製造を行うことができる。 In one embodiment, the preliminary confirmation can be performed as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can appropriately modify them to perform the preliminary confirmation manufacturing.
一つの実施形態では、事前の確認は以下のように行うことができる。以下に例示する各種数値、試薬、手順等は、代表例であり、当業者は適宜変更して事前の確認の製造を行うことができる。 In one embodiment, the preliminary confirmation can be performed as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can appropriately modify them to perform the preliminary confirmation manufacturing.
ドナー及び患者の感染症スクリーニング検査を実施し、ドナーについてはHBs抗原、HCV抗体、HIV-1/2、HTLV-1抗体が全て陰性であることを確認する。
Infectious disease screening tests are conducted on donors and patients, and donors are confirmed to be negative for HBs antigen, HCV antibody, HIV-1/2, and HTLV-1 antibody.
1.ドナーリンパ球の分離(無菌下で行う)
一つの実施形態では、ドナーリンパ球の分離は以下のように製造することができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更してドナーリンパ球の分離を行うことができる。
・アフェレーシスでドナーリンパ球を回収バッグに採取し、その回収バッグを放射線照射する。
・前述の放射線照射済み末梢血単核球を、適量のFicoll-Paque PREMIUM(GE Healthcare #17-5442-02)またはLymphocyte separation Solution(ナカライテスク#20828)等(例えば、20mL)が入った遠心管に入れ、860Gで20分間、22℃で遠心分離する。
・上清を捨て、リンパ球層を含む細胞浮遊液を、別の遠心管(例えば、50mL遠心管2本)に移す。
・細胞浮遊液の入った遠心管に、生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、シリンジ(例えば、18G注射針をつけた50mLシリンジ)またはピペットで吸引排出を繰り返して、よく混和させる。
・500Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を捨て、再度生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、細胞ペレットを、ピペットで吸引排出を繰り返して、よく混和させる。
・500Gで5分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を捨てる。
・予めドナーより採取した血漿を含むALyS505N-0培養液(細胞科学研究所 (CSTI)1020P10))を、細胞ペレットに加え(例えば、全液量が31mLになるまで適量)、ピペットで吸引排出を繰り返して、よく混和させる。
・シリンジ(例えば、18G注射針をつけた1mLシリンジ)またはピペットで、適量(例えば、0.3mL)抜き取り、細胞数および生細胞数を確認する。 1. Isolation of donor lymphocytes (performed under sterile conditions)
In one embodiment, donor lymphocytes can be separated as follows: The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can make appropriate modifications to separate donor lymphocytes.
- Donor lymphocytes are collected into a collection bag by apheresis, and the collection bag is then irradiated.
The irradiated peripheral blood mononuclear cells are placed in a centrifuge tube containing an appropriate amount of Ficoll-Paque PREMIUM (GE Healthcare #17-5442-02) or Lymphocyte Separation Solution (Nacalai Tesque #20828) or the like (e.g., 20 mL), and centrifuged at 860 G for 20 minutes at 22°C.
Discard the supernatant and transfer the cell suspension containing the lymphocyte layer to another centrifuge tube (for example, two 50 mL centrifuge tubes).
Add saline to the centrifuge tube containing the cell suspension (for example, an appropriate amount until the total volume is 50 mL), and mix well by repeatedly aspirating and dispensing with a syringe (for example, a 50 mL syringe with an 18G needle attached) or a pipette.
Centrifuge at 500G for 10 minutes at 22°C (you may set the centrifuge accelerator to fast and the brake to fast).
Discard the supernatant, add saline again (for example, an appropriate amount until the total volume is 50 mL), and mix the cell pellet well by repeatedly aspirating and dispensing the cell pellet with a pipette.
Centrifuge at 500G for 5 minutes at 22°C (you may set the centrifuge accelerator to fast and the brake to fast).
- Discard the supernatant.
Add ALyS505N-0 culture medium (Cell Science Institute (CSTI) 1020P10) containing plasma previously collected from a donor to the cell pellet (for example, an appropriate amount until the total volume is 31 mL), and mix well by repeatedly drawing in and out with a pipette.
Withdraw an appropriate amount (e.g., 0.3 mL) using a syringe (e.g., a 1 mL syringe with an 18G needle) or a pipette, and check the cell count and viable cell count.
一つの実施形態では、ドナーリンパ球の分離は以下のように製造することができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更してドナーリンパ球の分離を行うことができる。
・アフェレーシスでドナーリンパ球を回収バッグに採取し、その回収バッグを放射線照射する。
・前述の放射線照射済み末梢血単核球を、適量のFicoll-Paque PREMIUM(GE Healthcare #17-5442-02)またはLymphocyte separation Solution(ナカライテスク#20828)等(例えば、20mL)が入った遠心管に入れ、860Gで20分間、22℃で遠心分離する。
・上清を捨て、リンパ球層を含む細胞浮遊液を、別の遠心管(例えば、50mL遠心管2本)に移す。
・細胞浮遊液の入った遠心管に、生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、シリンジ(例えば、18G注射針をつけた50mLシリンジ)またはピペットで吸引排出を繰り返して、よく混和させる。
・500Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を捨て、再度生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、細胞ペレットを、ピペットで吸引排出を繰り返して、よく混和させる。
・500Gで5分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を捨てる。
・予めドナーより採取した血漿を含むALyS505N-0培養液(細胞科学研究所 (CSTI)1020P10))を、細胞ペレットに加え(例えば、全液量が31mLになるまで適量)、ピペットで吸引排出を繰り返して、よく混和させる。
・シリンジ(例えば、18G注射針をつけた1mLシリンジ)またはピペットで、適量(例えば、0.3mL)抜き取り、細胞数および生細胞数を確認する。 1. Isolation of donor lymphocytes (performed under sterile conditions)
In one embodiment, donor lymphocytes can be separated as follows: The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can make appropriate modifications to separate donor lymphocytes.
- Donor lymphocytes are collected into a collection bag by apheresis, and the collection bag is then irradiated.
The irradiated peripheral blood mononuclear cells are placed in a centrifuge tube containing an appropriate amount of Ficoll-Paque PREMIUM (GE Healthcare #17-5442-02) or Lymphocyte Separation Solution (Nacalai Tesque #20828) or the like (e.g., 20 mL), and centrifuged at 860 G for 20 minutes at 22°C.
Discard the supernatant and transfer the cell suspension containing the lymphocyte layer to another centrifuge tube (for example, two 50 mL centrifuge tubes).
Add saline to the centrifuge tube containing the cell suspension (for example, an appropriate amount until the total volume is 50 mL), and mix well by repeatedly aspirating and dispensing with a syringe (for example, a 50 mL syringe with an 18G needle attached) or a pipette.
Centrifuge at 500G for 10 minutes at 22°C (you may set the centrifuge accelerator to fast and the brake to fast).
Discard the supernatant, add saline again (for example, an appropriate amount until the total volume is 50 mL), and mix the cell pellet well by repeatedly aspirating and dispensing the cell pellet with a pipette.
Centrifuge at 500G for 5 minutes at 22°C (you may set the centrifuge accelerator to fast and the brake to fast).
- Discard the supernatant.
Add ALyS505N-0 culture medium (Cell Science Institute (CSTI) 1020P10) containing plasma previously collected from a donor to the cell pellet (for example, an appropriate amount until the total volume is 31 mL), and mix well by repeatedly drawing in and out with a pipette.
Withdraw an appropriate amount (e.g., 0.3 mL) using a syringe (e.g., a 1 mL syringe with an 18G needle) or a pipette, and check the cell count and viable cell count.
2.ドナーリンパ球の凍結保存(無菌下で行う)
一つの実施形態では、ドナーリンパ球の凍結保存は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更してドナーリンパ球の凍結保存を行うことができる。
・凍結バッグ(例えば、フローズバッグF-050 25mL凍結バッグ(株)ニプロ89-101)を無菌下で開封し、ラベルに必要事項(日付、製造番号、ドナー名)を記入する。
・シリンジ(例えば、18G注射針をつけた30mLシリンジ)で、細胞浮遊液を取り、凍結バッグに入れる。
・ACD液((株)テルモTP-A05ACD、例えば、細胞浮遊液15mLに対して2mL)を、細胞浮遊液が入った凍結バッグに加え、4℃で冷却した保冷剤に挟んで、10分間程度冷やす。
・シリンジ(例えば、18G注射針をつけた20mLシリンジ)を用いて、4℃で冷却したCP-1(極東製薬工業(株) 551-27202-4細胞凍害保護液CP-1)例えば、8.5mL)を、凍結バッグに1分半程度の時間をかけて加える。この際、凍結バッグをゆっくり攪拌する。
・シリンジを用いて、凍結バッグおよびそのポート内の空気を全て抜き取る。
・チューブシーラーを使用して凍結バッグをシールし、まず4℃で約5~10分冷却し、その後-80℃の冷凍庫で保管する。 2. Freezing and preserving donor lymphocytes (performed under sterile conditions)
In one embodiment, the cryopreservation of donor lymphocytes can be performed as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to cryopreserve donor lymphocytes.
- Open a freezing bag (for example, Frozebag F-050 25 mL freezing bag, Nipro 89-101) under aseptic conditions and write the necessary information (date, serial number, donor name) on the label.
Using a syringe (e.g., a 30 mL syringe fitted with an 18G needle), draw up the cell suspension and place it into a freezing bag.
Add ACD liquid (Terumo TP-A05ACD, for example, 2 mL per 15 mL of cell suspension) to the freezing bag containing the cell suspension, sandwich it between ice packs cooled to 4°C, and chill for about 10 minutes.
Using a syringe (e.g., a 20 mL syringe with an 18G needle), add CP-1 (Kyokuto Pharmaceutical Co., Ltd. 551-27202-4 Cell Cryoprotectant CP-1), e.g., 8.5 mL) cooled to 4°C to the freezing bag over a period of about one and a half minutes. During this time, slowly stir the freezing bag.
- Using a syringe, remove all air from the freezing bag and its ports.
Seal the freezing bag using a tube sealer and first chill at 4°C for approximately 5-10 minutes, then store in a -80°C freezer.
一つの実施形態では、ドナーリンパ球の凍結保存は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更してドナーリンパ球の凍結保存を行うことができる。
・凍結バッグ(例えば、フローズバッグF-050 25mL凍結バッグ(株)ニプロ89-101)を無菌下で開封し、ラベルに必要事項(日付、製造番号、ドナー名)を記入する。
・シリンジ(例えば、18G注射針をつけた30mLシリンジ)で、細胞浮遊液を取り、凍結バッグに入れる。
・ACD液((株)テルモTP-A05ACD、例えば、細胞浮遊液15mLに対して2mL)を、細胞浮遊液が入った凍結バッグに加え、4℃で冷却した保冷剤に挟んで、10分間程度冷やす。
・シリンジ(例えば、18G注射針をつけた20mLシリンジ)を用いて、4℃で冷却したCP-1(極東製薬工業(株) 551-27202-4細胞凍害保護液CP-1)例えば、8.5mL)を、凍結バッグに1分半程度の時間をかけて加える。この際、凍結バッグをゆっくり攪拌する。
・シリンジを用いて、凍結バッグおよびそのポート内の空気を全て抜き取る。
・チューブシーラーを使用して凍結バッグをシールし、まず4℃で約5~10分冷却し、その後-80℃の冷凍庫で保管する。 2. Freezing and preserving donor lymphocytes (performed under sterile conditions)
In one embodiment, the cryopreservation of donor lymphocytes can be performed as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to cryopreserve donor lymphocytes.
- Open a freezing bag (for example, Frozebag F-050 25 mL freezing bag, Nipro 89-101) under aseptic conditions and write the necessary information (date, serial number, donor name) on the label.
Using a syringe (e.g., a 30 mL syringe fitted with an 18G needle), draw up the cell suspension and place it into a freezing bag.
Add ACD liquid (Terumo TP-A05ACD, for example, 2 mL per 15 mL of cell suspension) to the freezing bag containing the cell suspension, sandwich it between ice packs cooled to 4°C, and chill for about 10 minutes.
Using a syringe (e.g., a 20 mL syringe with an 18G needle), add CP-1 (Kyokuto Pharmaceutical Co., Ltd. 551-27202-4 Cell Cryoprotectant CP-1), e.g., 8.5 mL) cooled to 4°C to the freezing bag over a period of about one and a half minutes. During this time, slowly stir the freezing bag.
- Using a syringe, remove all air from the freezing bag and its ports.
Seal the freezing bag using a tube sealer and first chill at 4°C for approximately 5-10 minutes, then store in a -80°C freezer.
3.ドナーリンパ球の解凍(無菌下で行う)
一つの実施形態では、ドナーリンパ球の解凍は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更してドナーリンパ球の解凍を行うことができる。
・保存されたドナー細胞の凍結バッグを、例えば、37℃恒温槽で解凍する。以降の操作は、好ましくは無菌下で行う。
・シリンジ(例えば、18G注射針をつけた50mLシリンジ)を用いて、解凍された凍結バッグから細胞浮遊液を抜き取り、遠心管(例えば、50mL遠心管2本に12.5mLずつ)に移す。
・細胞浮遊液の入った遠心管に、例えば5%アルブミン液(日本製薬(株) 123146364 献血アルブミン5%静注12.5g/250mL)(例えば、細胞浮遊液12.5mLに対して37.5mL)を追加し、よく混和させる。その後、約5分間静置する。
・例えば、600Gで10分間、22℃で遠心分離(例えば、好ましくは、遠心分離機のアクセルをfast、ブレーキをslowに設定する)する。
・上清を静かに捨て、細胞ペレットに、洗浄用のアルブミン加生理食塩液(例えば、5%アルブミン液25mLと生理食塩液19mLから作製する)等の適宜の液を加えて懸濁する。
・例えば、600Gで10分間、22℃で遠心分離(例えば、好ましくは、遠心分離機のアクセルをfast、ブレーキをslowに設定する)する。
・上清を静かに捨て、細胞ペレットに、ALyS505N培養液(例えば、50mL遠心管に対して10mL)を加えて懸濁する。
・ALyS505N-0培養液またはそれと同等の液が入った培養バッグ(例えば、(株)ニプロ87598 ニプロメディウムALyS505NB10)に、抗ヒトCD80抗体(例えば、m2D10.4;Cat.No.16-0809-85、eBioscience社)と抗ヒトCD86抗体(例えば、IT2.2;Cat.No.16-0869-85、eBioscience社)をそれぞれ例えば最終濃度10μg/mLで加え(またはCTLA4-Ig融合タンパク質(例えば、ベラタセプト)などの阻害因子を加え)、この培養バッグに、上記の細胞浮遊液をシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加える。一例では、培養バッグ中の総液量は、約840mLである。 3. Thawing donor lymphocytes (performed under sterile conditions)
In one embodiment, thawing of donor lymphocytes can be performed as follows: The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to thaw donor lymphocytes.
Thaw the frozen bag of stored donor cells, for example, in a constant temperature bath at 37° C. The subsequent operations are preferably performed under aseptic conditions.
Using a syringe (e.g., a 50 mL syringe with an 18G needle attached), withdraw the cell suspension from the thawed freezing bag and transfer it to a centrifuge tube (e.g., 12.5 mL each into two 50 mL centrifuge tubes).
Add, for example, 5% albumin solution (Nihon Pharmaceutical Co., Ltd. 123146364 Blood Donation Albumin 5% Intravenous Injection 12.5 g/250 mL) (for example, 37.5 mL per 12.5 mL of cell suspension) to the centrifuge tube containing the cell suspension and mix well. Then, leave to stand for about 5 minutes.
Centrifuge, for example, at 600G for 10 minutes at 22°C (for example, preferably with the centrifuge accelerator set to fast and the brake set to slow).
Carefully discard the supernatant, and add an appropriate liquid such as albumin-added saline for washing (for example, made from 25 mL of 5% albumin solution and 19 mL of saline) to the cell pellet to suspend it.
Centrifuge, for example, at 600G for 10 minutes at 22°C (for example, preferably with the centrifuge accelerator set to fast and the brake to slow).
Carefully discard the supernatant and add ALyS505N culture medium (for example, 10 mL for a 50 mL centrifuge tube) to the cell pellet to suspend it.
-Anti-human CD80 antibody (e.g., m2D10.4; Cat. No. 16-0809-85, eBioscience) and anti-human CD86 antibody (e.g., IT2.2; Cat. No. 16-0869-85, eBioscience) are added to a culture bag containing ALyS505N-0 culture medium or a liquid equivalent thereto (e.g., Nipro 87598 Nipro Medium ALyS505NB10) at a final concentration of, for example, 10 μg/mL (or an inhibitor such as a CTLA4-Ig fusion protein (e.g., belatacept) is added), and the above cell suspension is added to this culture bag by injection with a syringe (e.g., a 20 mL syringe with an 18G needle). In one example, the total volume of the liquid in the culture bag is about 840 mL.
一つの実施形態では、ドナーリンパ球の解凍は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更してドナーリンパ球の解凍を行うことができる。
・保存されたドナー細胞の凍結バッグを、例えば、37℃恒温槽で解凍する。以降の操作は、好ましくは無菌下で行う。
・シリンジ(例えば、18G注射針をつけた50mLシリンジ)を用いて、解凍された凍結バッグから細胞浮遊液を抜き取り、遠心管(例えば、50mL遠心管2本に12.5mLずつ)に移す。
・細胞浮遊液の入った遠心管に、例えば5%アルブミン液(日本製薬(株) 123146364 献血アルブミン5%静注12.5g/250mL)(例えば、細胞浮遊液12.5mLに対して37.5mL)を追加し、よく混和させる。その後、約5分間静置する。
・例えば、600Gで10分間、22℃で遠心分離(例えば、好ましくは、遠心分離機のアクセルをfast、ブレーキをslowに設定する)する。
・上清を静かに捨て、細胞ペレットに、洗浄用のアルブミン加生理食塩液(例えば、5%アルブミン液25mLと生理食塩液19mLから作製する)等の適宜の液を加えて懸濁する。
・例えば、600Gで10分間、22℃で遠心分離(例えば、好ましくは、遠心分離機のアクセルをfast、ブレーキをslowに設定する)する。
・上清を静かに捨て、細胞ペレットに、ALyS505N培養液(例えば、50mL遠心管に対して10mL)を加えて懸濁する。
・ALyS505N-0培養液またはそれと同等の液が入った培養バッグ(例えば、(株)ニプロ87598 ニプロメディウムALyS505NB10)に、抗ヒトCD80抗体(例えば、m2D10.4;Cat.No.16-0809-85、eBioscience社)と抗ヒトCD86抗体(例えば、IT2.2;Cat.No.16-0869-85、eBioscience社)をそれぞれ例えば最終濃度10μg/mLで加え(またはCTLA4-Ig融合タンパク質(例えば、ベラタセプト)などの阻害因子を加え)、この培養バッグに、上記の細胞浮遊液をシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加える。一例では、培養バッグ中の総液量は、約840mLである。 3. Thawing donor lymphocytes (performed under sterile conditions)
In one embodiment, thawing of donor lymphocytes can be performed as follows: The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to thaw donor lymphocytes.
Thaw the frozen bag of stored donor cells, for example, in a constant temperature bath at 37° C. The subsequent operations are preferably performed under aseptic conditions.
Using a syringe (e.g., a 50 mL syringe with an 18G needle attached), withdraw the cell suspension from the thawed freezing bag and transfer it to a centrifuge tube (e.g., 12.5 mL each into two 50 mL centrifuge tubes).
Add, for example, 5% albumin solution (Nihon Pharmaceutical Co., Ltd. 123146364 Blood Donation Albumin 5% Intravenous Injection 12.5 g/250 mL) (for example, 37.5 mL per 12.5 mL of cell suspension) to the centrifuge tube containing the cell suspension and mix well. Then, leave to stand for about 5 minutes.
Centrifuge, for example, at 600G for 10 minutes at 22°C (for example, preferably with the centrifuge accelerator set to fast and the brake set to slow).
Carefully discard the supernatant, and add an appropriate liquid such as albumin-added saline for washing (for example, made from 25 mL of 5% albumin solution and 19 mL of saline) to the cell pellet to suspend it.
Centrifuge, for example, at 600G for 10 minutes at 22°C (for example, preferably with the centrifuge accelerator set to fast and the brake to slow).
Carefully discard the supernatant and add ALyS505N culture medium (for example, 10 mL for a 50 mL centrifuge tube) to the cell pellet to suspend it.
-Anti-human CD80 antibody (e.g., m2D10.4; Cat. No. 16-0809-85, eBioscience) and anti-human CD86 antibody (e.g., IT2.2; Cat. No. 16-0869-85, eBioscience) are added to a culture bag containing ALyS505N-0 culture medium or a liquid equivalent thereto (e.g., Nipro 87598 Nipro Medium ALyS505NB10) at a final concentration of, for example, 10 μg/mL (or an inhibitor such as a CTLA4-Ig fusion protein (e.g., belatacept) is added), and the above cell suspension is added to this culture bag by injection with a syringe (e.g., a 20 mL syringe with an 18G needle). In one example, the total volume of the liquid in the culture bag is about 840 mL.
4.患者リンパ球の分離~一次培養開始(無菌下で行う)
一つの実施形態では、患者リンパ球の分離は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更して患者リンパ球の分離を行うことができる。
・患者より採取した血漿は、恒温槽中、例えば、56℃で30分間加温し、非働化しておく。直ちに使用しないものは、凍結保存する。
・患者より採取した末梢血を、適量の適切な媒体、例えばFicoll-Paque(例えば、20mL)が入った遠心管に入れ、例えば、860Gで20分間、22℃で遠心分離(例えば、好ましくは、遠心分離機のアクセルをslow、ブレーキをslowに設定する)する。
・上清を捨て、リンパ球層を含む細胞浮遊液を、別の遠心管(例えば、50mL遠心管2本)に移す。
・細胞浮遊液の入った遠心管に、生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、ピペットで吸引排出を繰り返して、よく混和させる。
・例えば、500Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を捨て、再度生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、細胞ペレットを、ピペットで吸引排出を繰り返して、よく混和させる。
・例えば、500Gで5分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を捨て、細胞ペレットに、例えば、ALyS505N-0培養液(例えば、10mL)を加えて懸濁し、細胞浮遊液を作製する(例えば、合計20mLになるまでALyS505N-0培養液を追加する)。ここで、0.5mL程度の細胞浮遊液を抜き取り、細胞数、生細胞数および表面抗原の発現を確認する。
・「3.ドナーリンパ球の解凍」で作製したALyS505N-0培養液中ドナー細胞および抗体等の阻害因子が入った培養バッグに患者由来の非働化血漿を追加する。
・この培養バッグに、上記患者由来細胞浮遊液をシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加え、チューブシーラーを使用して培養バッグをシールする。一例では、培養バッグ中の総液量は、約1000mLである。
・37℃インキュベーター内で、例えば、1週間培養する。 4. Isolation of patient lymphocytes - starting primary culture (performed under sterile conditions)
In one embodiment, the patient's lymphocytes can be separated as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to separate the patient's lymphocytes.
Plasma collected from the patient is inactivated by heating it in a constant temperature bath, for example, at 56°C for 30 minutes. If it is not used immediately, it is stored frozen.
Peripheral blood collected from a patient is placed in a centrifuge tube containing an appropriate amount of a suitable medium, such as Ficoll-Paque (e.g., 20 mL), and centrifuged, for example, at 860 G for 20 minutes at 22°C (e.g., preferably with the accelerator and brake of the centrifuge set to slow).
Discard the supernatant and transfer the cell suspension containing the lymphocyte layer to another centrifuge tube (for example, two 50 mL centrifuge tubes).
Add saline to the centrifuge tube containing the cell suspension (for example, an appropriate amount until the total volume is 50 mL), and mix well by repeatedly aspirating and dispensing with a pipette.
For example, centrifuge at 500 G for 10 minutes at 22° C. (the accelerator and brake of the centrifuge may be set to fast and fast, respectively).
Discard the supernatant, add saline again (for example, an appropriate amount until the total volume is 50 mL), and mix the cell pellet well by repeatedly aspirating and dispensing the cell pellet with a pipette.
For example, centrifuge at 500 G for 5 minutes at 22° C. (the accelerator and brake of the centrifuge may be set to fast and fast, respectively).
The supernatant is discarded, and the cell pellet is suspended in, for example, ALyS505N-0 culture medium (for example, 10 mL) to prepare a cell suspension (for example, add ALyS505N-0 culture medium until the total volume becomes 20 mL). At this point, about 0.5 mL of the cell suspension is withdrawn, and the cell count, viable cell count, and surface antigen expression are confirmed.
- Inactivated plasma derived from the patient is added to the culture bag containing the donor cells in the ALyS505N-0 culture medium prepared in "3. Thawing of donor lymphocytes" and inhibitors such as antibodies.
The patient-derived cell suspension is added to the culture bag by injection using a syringe (e.g., a 20 mL syringe with an 18G needle) and the culture bag is sealed using a tube sealer. In one example, the total volume of the culture bag is about 1000 mL.
- Culture in a 37°C incubator for, for example, one week.
一つの実施形態では、患者リンパ球の分離は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更して患者リンパ球の分離を行うことができる。
・患者より採取した血漿は、恒温槽中、例えば、56℃で30分間加温し、非働化しておく。直ちに使用しないものは、凍結保存する。
・患者より採取した末梢血を、適量の適切な媒体、例えばFicoll-Paque(例えば、20mL)が入った遠心管に入れ、例えば、860Gで20分間、22℃で遠心分離(例えば、好ましくは、遠心分離機のアクセルをslow、ブレーキをslowに設定する)する。
・上清を捨て、リンパ球層を含む細胞浮遊液を、別の遠心管(例えば、50mL遠心管2本)に移す。
・細胞浮遊液の入った遠心管に、生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、ピペットで吸引排出を繰り返して、よく混和させる。
・例えば、500Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を捨て、再度生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、細胞ペレットを、ピペットで吸引排出を繰り返して、よく混和させる。
・例えば、500Gで5分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を捨て、細胞ペレットに、例えば、ALyS505N-0培養液(例えば、10mL)を加えて懸濁し、細胞浮遊液を作製する(例えば、合計20mLになるまでALyS505N-0培養液を追加する)。ここで、0.5mL程度の細胞浮遊液を抜き取り、細胞数、生細胞数および表面抗原の発現を確認する。
・「3.ドナーリンパ球の解凍」で作製したALyS505N-0培養液中ドナー細胞および抗体等の阻害因子が入った培養バッグに患者由来の非働化血漿を追加する。
・この培養バッグに、上記患者由来細胞浮遊液をシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加え、チューブシーラーを使用して培養バッグをシールする。一例では、培養バッグ中の総液量は、約1000mLである。
・37℃インキュベーター内で、例えば、1週間培養する。 4. Isolation of patient lymphocytes - starting primary culture (performed under sterile conditions)
In one embodiment, the patient's lymphocytes can be separated as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to separate the patient's lymphocytes.
Plasma collected from the patient is inactivated by heating it in a constant temperature bath, for example, at 56°C for 30 minutes. If it is not used immediately, it is stored frozen.
Peripheral blood collected from a patient is placed in a centrifuge tube containing an appropriate amount of a suitable medium, such as Ficoll-Paque (e.g., 20 mL), and centrifuged, for example, at 860 G for 20 minutes at 22°C (e.g., preferably with the accelerator and brake of the centrifuge set to slow).
Discard the supernatant and transfer the cell suspension containing the lymphocyte layer to another centrifuge tube (for example, two 50 mL centrifuge tubes).
Add saline to the centrifuge tube containing the cell suspension (for example, an appropriate amount until the total volume is 50 mL), and mix well by repeatedly aspirating and dispensing with a pipette.
For example, centrifuge at 500 G for 10 minutes at 22° C. (the accelerator and brake of the centrifuge may be set to fast and fast, respectively).
Discard the supernatant, add saline again (for example, an appropriate amount until the total volume is 50 mL), and mix the cell pellet well by repeatedly aspirating and dispensing the cell pellet with a pipette.
For example, centrifuge at 500 G for 5 minutes at 22° C. (the accelerator and brake of the centrifuge may be set to fast and fast, respectively).
The supernatant is discarded, and the cell pellet is suspended in, for example, ALyS505N-0 culture medium (for example, 10 mL) to prepare a cell suspension (for example, add ALyS505N-0 culture medium until the total volume becomes 20 mL). At this point, about 0.5 mL of the cell suspension is withdrawn, and the cell count, viable cell count, and surface antigen expression are confirmed.
- Inactivated plasma derived from the patient is added to the culture bag containing the donor cells in the ALyS505N-0 culture medium prepared in "3. Thawing of donor lymphocytes" and inhibitors such as antibodies.
The patient-derived cell suspension is added to the culture bag by injection using a syringe (e.g., a 20 mL syringe with an 18G needle) and the culture bag is sealed using a tube sealer. In one example, the total volume of the culture bag is about 1000 mL.
- Culture in a 37°C incubator for, for example, one week.
5-1.培地交換(例えば、1週間目、好ましくは無菌下で行う)
一つの実施形態では、培地交換は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更して培地交換を行うことができる。
・インキュベーターから培養バッグを取り出し、内容物を、遠心管(例えば、225mL遠心管4本)に分注する。
・例えば、600Gで10分間、22℃で遠心分離(例えば、好ましくは、遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を静かに捨て、細胞ペレットに、例えば、ALyS505N-0培養液を加えて懸濁し、細胞浮遊液を作製する(例えば、合計20mLになるまでALyS505N-0培養液を追加する)。ここで、0.3mL程度の細胞浮遊液を抜き取り、細胞数および生細胞数を確認する。
・例えば、ALyS505N-0培養液が入った培養バッグに、細胞浮遊液をシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加える。
・抗ヒトCD80抗体(例えば、2D10.4)希釈液と抗ヒトCD86抗体(例えば、IT2.2)希釈液を、それぞれ例えば、最終濃度10μg/mLとなるように(またはCTLA4-Ig融合タンパク質(例えば、ベラタセプト)等の阻害因子を)、培養バッグにシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加える。 5-1. Medium exchange (e.g., after 1 week, preferably under sterile conditions)
In one embodiment, the medium exchange can be performed as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and a person skilled in the art can appropriately modify them to exchange the medium.
- Remove the culture bag from the incubator and dispense the contents into centrifuge tubes (e.g., four 225 mL centrifuge tubes).
Centrifuge, for example, at 600G for 10 minutes at 22°C (for example, preferably with the centrifuge accelerator set to fast and the brake set to fast).
The supernatant is gently discarded, and the cell pellet is suspended in, for example, ALyS505N-0 culture medium to prepare a cell suspension (for example, add ALyS505N-0 culture medium until the total volume becomes 20 mL). At this point, about 0.3 mL of the cell suspension is removed and the cell count and viable cell count are confirmed.
For example, the cell suspension is added to a culture bag containing ALyS505N-0 culture medium by injection using a syringe (for example, a 20 mL syringe with an 18G needle).
- Add a diluted solution of anti-human CD80 antibody (e.g., 2D10.4) and an diluted solution of anti-human CD86 antibody (e.g., IT2.2) to a final concentration of, for example, 10 μg/mL (or an inhibitor such as a CTLA4-Ig fusion protein (e.g., belatacept)) to the culture bag by injecting with a syringe (e.g., a 20 mL syringe attached with an 18G needle).
一つの実施形態では、培地交換は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更して培地交換を行うことができる。
・インキュベーターから培養バッグを取り出し、内容物を、遠心管(例えば、225mL遠心管4本)に分注する。
・例えば、600Gで10分間、22℃で遠心分離(例えば、好ましくは、遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を静かに捨て、細胞ペレットに、例えば、ALyS505N-0培養液を加えて懸濁し、細胞浮遊液を作製する(例えば、合計20mLになるまでALyS505N-0培養液を追加する)。ここで、0.3mL程度の細胞浮遊液を抜き取り、細胞数および生細胞数を確認する。
・例えば、ALyS505N-0培養液が入った培養バッグに、細胞浮遊液をシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加える。
・抗ヒトCD80抗体(例えば、2D10.4)希釈液と抗ヒトCD86抗体(例えば、IT2.2)希釈液を、それぞれ例えば、最終濃度10μg/mLとなるように(またはCTLA4-Ig融合タンパク質(例えば、ベラタセプト)等の阻害因子を)、培養バッグにシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加える。 5-1. Medium exchange (e.g., after 1 week, preferably under sterile conditions)
In one embodiment, the medium exchange can be performed as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and a person skilled in the art can appropriately modify them to exchange the medium.
- Remove the culture bag from the incubator and dispense the contents into centrifuge tubes (e.g., four 225 mL centrifuge tubes).
Centrifuge, for example, at 600G for 10 minutes at 22°C (for example, preferably with the centrifuge accelerator set to fast and the brake set to fast).
The supernatant is gently discarded, and the cell pellet is suspended in, for example, ALyS505N-0 culture medium to prepare a cell suspension (for example, add ALyS505N-0 culture medium until the total volume becomes 20 mL). At this point, about 0.3 mL of the cell suspension is removed and the cell count and viable cell count are confirmed.
For example, the cell suspension is added to a culture bag containing ALyS505N-0 culture medium by injection using a syringe (for example, a 20 mL syringe with an 18G needle).
- Add a diluted solution of anti-human CD80 antibody (e.g., 2D10.4) and an diluted solution of anti-human CD86 antibody (e.g., IT2.2) to a final concentration of, for example, 10 μg/mL (or an inhibitor such as a CTLA4-Ig fusion protein (e.g., belatacept)) to the culture bag by injecting with a syringe (e.g., a 20 mL syringe attached with an 18G needle).
5-2.ドナーリンパ球の解凍
・抗原再刺激~二次培養開始(例えば、1週間目、無菌下で行う) 一つの実施形態では、ドナーリンパ球の解凍
・抗原再刺激~二次培養開始は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更してドナーリンパ球の解凍
・抗原再刺激~二次培養開始を行うことができる。
・保存されたドナー細胞の凍結バッグと患者由来の非働化血漿を、例えば、37℃恒温槽で解凍する。以降の操作は、好ましくは、無菌下で行う。
・シリンジ(例えば、18G注射針をつけた50mLシリンジ)を用いて、解凍された凍結バッグからドナー細胞浮遊液を抜き取り、遠心管(例えば、50mL遠心管2本)に移す。
・ドナー細胞浮遊液の入った遠心管に、5%アルブミン液(例えば、50mL遠心管2本につき、合計で約50mL)を追加し、よく混和させる。その後、約5分間静置する。
・例えば、600Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをslowに設定する)する。
・上清を静かに捨て、細胞ペレットに、洗浄用のアルブミン加生理食塩液(例えば、5%アルブミン液25mLと生理食塩液19mLから作製する)を加えて懸濁する。
・例えば、600Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをslowに設定する)する。
・上清を静かに捨て、細胞ペレットに、例えば、ALyS505N-0培養液(例えば、50mL遠心管に対して10mL)を加えて懸濁する。
・「3.ドナーリンパ球の解凍」で作製したALyS505N培養液中患者細胞および抗体等の阻害因子が入った培養バッグに、解凍した患者由来の非働化血漿(例えば、10mL)をシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加え、さらに、この培養バッグに、上記の細胞浮遊液をシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加える。一例では、培養バッグ中の総液量は、約1000mLである。
・チューブシーラーを使用して培養バッグをシールする。
・37℃インキュベーター内で、例えば、1週間培養する。 5-2. Thawing of donor lymphocytes, antigen restimulation, and initiation of secondary culture (for example, in the first week, under aseptic conditions) In one embodiment, thawing of donor lymphocytes, antigen restimulation, and initiation of secondary culture can be performed as follows. The various numerical values, reagents, procedures, etc. exemplified below are representative examples, and a person skilled in the art can appropriately modify the thawing of donor lymphocytes, antigen restimulation, and initiation of secondary culture.
The frozen bag of stored donor cells and the inactivated plasma from the patient are thawed, for example, in a thermostatic bath at 37° C. The subsequent operations are preferably performed under aseptic conditions.
Using a syringe (e.g., a 50 mL syringe with an 18G needle), withdraw the donor cell suspension from the thawed cryobag and transfer it to a centrifuge tube (e.g., two 50 mL centrifuge tubes).
Add 5% albumin solution (for example, about 50 mL in total for two 50 mL centrifuge tubes) to the centrifuge tube containing the donor cell suspension, mix well, and then leave to stand for about 5 minutes.
For example, centrifuge at 600 G for 10 minutes at 22° C. (set the centrifuge's accelerator to fast and the brake to slow).
Carefully discard the supernatant, and add albumin-containing saline for washing (prepared, for example, from 25 mL of 5% albumin solution and 19 mL of saline) to the cell pellet to suspend it.
For example, centrifuge at 600 G for 10 minutes at 22° C. (set the centrifuge's accelerator to fast and the brake to slow).
Carefully discard the supernatant and suspend the cell pellet in, for example, ALyS505N-0 culture medium (for example, 10 mL for a 50 mL centrifuge tube).
- In the culture bag containing the patient cells in the ALyS505N culture medium and inhibitors such as antibodies prepared in "3. Thawing of donor lymphocytes", the thawed inactivated plasma from the patient (e.g., 10 mL) is added by injection with a syringe (e.g., a 20 mL syringe with an 18 G needle), and further, the above-mentioned cell suspension is added by injection with a syringe (e.g., a 20 mL syringe with an 18 G needle) to this culture bag. In one example, the total volume of the liquid in the culture bag is about 1000 mL.
- Seal the culture bag using a tube sealer.
- Culture in a 37°C incubator for, for example, one week.
・抗原再刺激~二次培養開始(例えば、1週間目、無菌下で行う) 一つの実施形態では、ドナーリンパ球の解凍
・抗原再刺激~二次培養開始は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更してドナーリンパ球の解凍
・抗原再刺激~二次培養開始を行うことができる。
・保存されたドナー細胞の凍結バッグと患者由来の非働化血漿を、例えば、37℃恒温槽で解凍する。以降の操作は、好ましくは、無菌下で行う。
・シリンジ(例えば、18G注射針をつけた50mLシリンジ)を用いて、解凍された凍結バッグからドナー細胞浮遊液を抜き取り、遠心管(例えば、50mL遠心管2本)に移す。
・ドナー細胞浮遊液の入った遠心管に、5%アルブミン液(例えば、50mL遠心管2本につき、合計で約50mL)を追加し、よく混和させる。その後、約5分間静置する。
・例えば、600Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをslowに設定する)する。
・上清を静かに捨て、細胞ペレットに、洗浄用のアルブミン加生理食塩液(例えば、5%アルブミン液25mLと生理食塩液19mLから作製する)を加えて懸濁する。
・例えば、600Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをslowに設定する)する。
・上清を静かに捨て、細胞ペレットに、例えば、ALyS505N-0培養液(例えば、50mL遠心管に対して10mL)を加えて懸濁する。
・「3.ドナーリンパ球の解凍」で作製したALyS505N培養液中患者細胞および抗体等の阻害因子が入った培養バッグに、解凍した患者由来の非働化血漿(例えば、10mL)をシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加え、さらに、この培養バッグに、上記の細胞浮遊液をシリンジ(例えば、18G注射針をつけた20mLシリンジ)で注入して加える。一例では、培養バッグ中の総液量は、約1000mLである。
・チューブシーラーを使用して培養バッグをシールする。
・37℃インキュベーター内で、例えば、1週間培養する。 5-2. Thawing of donor lymphocytes, antigen restimulation, and initiation of secondary culture (for example, in the first week, under aseptic conditions) In one embodiment, thawing of donor lymphocytes, antigen restimulation, and initiation of secondary culture can be performed as follows. The various numerical values, reagents, procedures, etc. exemplified below are representative examples, and a person skilled in the art can appropriately modify the thawing of donor lymphocytes, antigen restimulation, and initiation of secondary culture.
The frozen bag of stored donor cells and the inactivated plasma from the patient are thawed, for example, in a thermostatic bath at 37° C. The subsequent operations are preferably performed under aseptic conditions.
Using a syringe (e.g., a 50 mL syringe with an 18G needle), withdraw the donor cell suspension from the thawed cryobag and transfer it to a centrifuge tube (e.g., two 50 mL centrifuge tubes).
Add 5% albumin solution (for example, about 50 mL in total for two 50 mL centrifuge tubes) to the centrifuge tube containing the donor cell suspension, mix well, and then leave to stand for about 5 minutes.
For example, centrifuge at 600 G for 10 minutes at 22° C. (set the centrifuge's accelerator to fast and the brake to slow).
Carefully discard the supernatant, and add albumin-containing saline for washing (prepared, for example, from 25 mL of 5% albumin solution and 19 mL of saline) to the cell pellet to suspend it.
For example, centrifuge at 600 G for 10 minutes at 22° C. (set the centrifuge's accelerator to fast and the brake to slow).
Carefully discard the supernatant and suspend the cell pellet in, for example, ALyS505N-0 culture medium (for example, 10 mL for a 50 mL centrifuge tube).
- In the culture bag containing the patient cells in the ALyS505N culture medium and inhibitors such as antibodies prepared in "3. Thawing of donor lymphocytes", the thawed inactivated plasma from the patient (e.g., 10 mL) is added by injection with a syringe (e.g., a 20 mL syringe with an 18 G needle), and further, the above-mentioned cell suspension is added by injection with a syringe (e.g., a 20 mL syringe with an 18 G needle) to this culture bag. In one example, the total volume of the liquid in the culture bag is about 1000 mL.
- Seal the culture bag using a tube sealer.
- Culture in a 37°C incubator for, for example, one week.
6.二次培養中の検査(培養細胞抜き取り試験)
一つの実施形態では、二次培養中の検査は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更して二次培養中の検査を行うことができる。
・代表的に、二次培養開始から3日目(培養通算10日目)に少量の培養液を、培養バッグから抜き取り、マイコプラズマ汚染などについて検査する。 6. Inspection during secondary culture (sampling test of cultured cells)
In one embodiment, the test during the secondary culture can be performed as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and a person skilled in the art can appropriately modify them to perform the test during the secondary culture.
Typically, on the third day after the start of the secondary culture (the 10th day of culture), a small amount of culture medium is withdrawn from the culture bag and tested for mycoplasma contamination, etc.
一つの実施形態では、二次培養中の検査は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更して二次培養中の検査を行うことができる。
・代表的に、二次培養開始から3日目(培養通算10日目)に少量の培養液を、培養バッグから抜き取り、マイコプラズマ汚染などについて検査する。 6. Inspection during secondary culture (sampling test of cultured cells)
In one embodiment, the test during the secondary culture can be performed as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and a person skilled in the art can appropriately modify them to perform the test during the secondary culture.
Typically, on the third day after the start of the secondary culture (the 10th day of culture), a small amount of culture medium is withdrawn from the culture bag and tested for mycoplasma contamination, etc.
7.培養リンパ球の回収・充填(無菌下で行う)
一つの実施形態では、培養リンパ球の回収
・充填は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更して培養リンパ球の回収
・充填を行うことができる。
・例えば、二次培養開始から7日目(培養通算14日目)インキュベーターから培養バッグを取り出し、内容物を、遠心管(例えば、225mL遠心管4本)に分注する。
・例えば、600Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を静かに捨て、細胞ペレットに、生理食塩液を加えて懸濁する。
・例えば、600Gで10分間、22℃で遠心分離(例えば、好ましくは、遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を静かに捨て、細胞ペレットに、生理食塩液(例えば、10mL)を加えて懸濁し、細胞浮遊液を作製する。
・細胞浮遊液を、適量のFicoll-Paque(例えば、20mL)が入った遠心管(例えば、50mL遠心管)に静かに入れて重層させる。
・例えば、860Gで20分間、22℃で遠心分離(遠心分離機のアクセルをslow、ブレーキをslowに設定する)する。
・上清を捨て、リンパ球層を含む細胞浮遊液を、別の遠心管(例えば、50mL遠心管)に移す。
・細胞浮遊液の入った遠心管に、生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、シリンジ(例えば、18G注射針をつけた50mLシリンジ)で吸引排出を繰り返して、よく混和させる。
・例えば、500Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を5mL程度残して他は捨て、ピペットで吸引排出を繰り返して、よく混和させる。
・生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、シリンジ(例えば、例えば、18G注射針をつけた50mLシリンジ)で吸引排出を繰り返して、よく混和させる(a)。
・例えば、500Gで5分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する(b)。
・上清を5mL程度残して他は捨て、ピペットで吸引排出を繰り返して、よく混和させる(c)。
・上記(a)、(b)および(c)をさらに2回繰り返す。
・最後の遠心分離後の上清を、適量(例えば、4mL)を抜き取り、無菌検査およびマイコプラズマ検査に供する。
・再度生理食塩液を加えて懸濁し、細胞浮遊液を、最終的な容器(例えば、100mL生理食塩液のボトル)に移す。適量(例えば、4mL)を抜き取り、最終的な生成物の、細胞数、生細胞数、表面抗原の発現およびエンドトキシンの含有量を確認する。 7. Collection and filling of cultured lymphocytes (performed under sterile conditions)
In one embodiment, the collection and filling of cultured lymphocytes can be performed as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to collect and fill cultured lymphocytes.
For example, on the 7th day after the start of the secondary culture (the 14th day in total), remove the culture bag from the incubator and dispense the contents into centrifuge tubes (for example, four 225 mL centrifuge tubes).
For example, centrifuge at 600 G for 10 minutes at 22° C. (the accelerator and brake of the centrifuge may be set to fast and fast, respectively).
- Carefully discard the supernatant and add saline to the cell pellet to suspend it.
Centrifuge, for example, at 600G for 10 minutes at 22°C (for example, preferably with the centrifuge accelerator set to fast and the brake set to fast).
Carefully discard the supernatant, and add physiological saline (for example, 10 mL) to the cell pellet to suspend it, thereby preparing a cell suspension.
The cell suspension is gently placed into a centrifuge tube (e.g., a 50 mL centrifuge tube) containing an appropriate amount of Ficoll-Paque (e.g., 20 mL) and layered.
For example, centrifuge at 860 G for 20 minutes at 22° C. (set the centrifuge accelerator to slow and the brake to slow).
Discard the supernatant and transfer the cell suspension containing the lymphocyte layer to another centrifuge tube (e.g., a 50 mL centrifuge tube).
Add saline to the centrifuge tube containing the cell suspension (for example, an appropriate amount until the total volume is 50 mL), and mix well by repeatedly aspirating and discharging with a syringe (for example, a 50 mL syringe with an 18G needle attached).
For example, centrifuge at 500 G for 10 minutes at 22° C. (the accelerator and brake of the centrifuge may be set to fast and fast, respectively).
- Discard all but 5 mL of the supernatant, and mix thoroughly by repeatedly aspirating and dispensing with a pipette.
Add saline (for example, an appropriate amount until the total volume is 50 mL) and mix well by repeatedly aspirating and discharging with a syringe (for example, a 50 mL syringe with an 18G needle) (a).
For example, centrifuge at 500 G for 5 minutes at 22° C. (the accelerator and brake of the centrifuge may be set to fast) (b).
Discard all but 5 mL of the supernatant, and mix thoroughly by repeatedly aspirating and dispensing with a pipette (c).
Repeat steps (a), (b) and (c) above two more times.
- After the final centrifugation, an appropriate amount (e.g., 4 mL) of the supernatant is withdrawn and subjected to sterility and mycoplasma testing.
Physiological saline is added again to suspend the cells, and the cell suspension is transferred to a final container (e.g., a 100 mL bottle of physiological saline). An appropriate amount (e.g., 4 mL) is withdrawn, and the cell count, viable cell count, surface antigen expression, and endotoxin content of the final product are confirmed.
一つの実施形態では、培養リンパ球の回収
・充填は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更して培養リンパ球の回収
・充填を行うことができる。
・例えば、二次培養開始から7日目(培養通算14日目)インキュベーターから培養バッグを取り出し、内容物を、遠心管(例えば、225mL遠心管4本)に分注する。
・例えば、600Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を静かに捨て、細胞ペレットに、生理食塩液を加えて懸濁する。
・例えば、600Gで10分間、22℃で遠心分離(例えば、好ましくは、遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を静かに捨て、細胞ペレットに、生理食塩液(例えば、10mL)を加えて懸濁し、細胞浮遊液を作製する。
・細胞浮遊液を、適量のFicoll-Paque(例えば、20mL)が入った遠心管(例えば、50mL遠心管)に静かに入れて重層させる。
・例えば、860Gで20分間、22℃で遠心分離(遠心分離機のアクセルをslow、ブレーキをslowに設定する)する。
・上清を捨て、リンパ球層を含む細胞浮遊液を、別の遠心管(例えば、50mL遠心管)に移す。
・細胞浮遊液の入った遠心管に、生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、シリンジ(例えば、18G注射針をつけた50mLシリンジ)で吸引排出を繰り返して、よく混和させる。
・例えば、500Gで10分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する。
・上清を5mL程度残して他は捨て、ピペットで吸引排出を繰り返して、よく混和させる。
・生理食塩液を追加し(例えば、全液量が50mLになるまで適量)、シリンジ(例えば、例えば、18G注射針をつけた50mLシリンジ)で吸引排出を繰り返して、よく混和させる(a)。
・例えば、500Gで5分間、22℃で遠心分離(遠心分離機のアクセルをfast、ブレーキをfastに設定してもよい)する(b)。
・上清を5mL程度残して他は捨て、ピペットで吸引排出を繰り返して、よく混和させる(c)。
・上記(a)、(b)および(c)をさらに2回繰り返す。
・最後の遠心分離後の上清を、適量(例えば、4mL)を抜き取り、無菌検査およびマイコプラズマ検査に供する。
・再度生理食塩液を加えて懸濁し、細胞浮遊液を、最終的な容器(例えば、100mL生理食塩液のボトル)に移す。適量(例えば、4mL)を抜き取り、最終的な生成物の、細胞数、生細胞数、表面抗原の発現およびエンドトキシンの含有量を確認する。 7. Collection and filling of cultured lymphocytes (performed under sterile conditions)
In one embodiment, the collection and filling of cultured lymphocytes can be performed as follows. The various values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can modify them as appropriate to collect and fill cultured lymphocytes.
For example, on the 7th day after the start of the secondary culture (the 14th day in total), remove the culture bag from the incubator and dispense the contents into centrifuge tubes (for example, four 225 mL centrifuge tubes).
For example, centrifuge at 600 G for 10 minutes at 22° C. (the accelerator and brake of the centrifuge may be set to fast and fast, respectively).
- Carefully discard the supernatant and add saline to the cell pellet to suspend it.
Centrifuge, for example, at 600G for 10 minutes at 22°C (for example, preferably with the centrifuge accelerator set to fast and the brake set to fast).
Carefully discard the supernatant, and add physiological saline (for example, 10 mL) to the cell pellet to suspend it, thereby preparing a cell suspension.
The cell suspension is gently placed into a centrifuge tube (e.g., a 50 mL centrifuge tube) containing an appropriate amount of Ficoll-Paque (e.g., 20 mL) and layered.
For example, centrifuge at 860 G for 20 minutes at 22° C. (set the centrifuge accelerator to slow and the brake to slow).
Discard the supernatant and transfer the cell suspension containing the lymphocyte layer to another centrifuge tube (e.g., a 50 mL centrifuge tube).
Add saline to the centrifuge tube containing the cell suspension (for example, an appropriate amount until the total volume is 50 mL), and mix well by repeatedly aspirating and discharging with a syringe (for example, a 50 mL syringe with an 18G needle attached).
For example, centrifuge at 500 G for 10 minutes at 22° C. (the accelerator and brake of the centrifuge may be set to fast and fast, respectively).
- Discard all but 5 mL of the supernatant, and mix thoroughly by repeatedly aspirating and dispensing with a pipette.
Add saline (for example, an appropriate amount until the total volume is 50 mL) and mix well by repeatedly aspirating and discharging with a syringe (for example, a 50 mL syringe with an 18G needle) (a).
For example, centrifuge at 500 G for 5 minutes at 22° C. (the accelerator and brake of the centrifuge may be set to fast) (b).
Discard all but 5 mL of the supernatant, and mix thoroughly by repeatedly aspirating and dispensing with a pipette (c).
Repeat steps (a), (b) and (c) above two more times.
- After the final centrifugation, an appropriate amount (e.g., 4 mL) of the supernatant is withdrawn and subjected to sterility and mycoplasma testing.
Physiological saline is added again to suspend the cells, and the cell suspension is transferred to a final container (e.g., a 100 mL bottle of physiological saline). An appropriate amount (e.g., 4 mL) is withdrawn, and the cell count, viable cell count, surface antigen expression, and endotoxin content of the final product are confirmed.
8.二次包装
一つの実施形態では、二次包装は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更して二次包装を行うことができる。
・代表的には、適切な基準(代表的には、NUHCPC-M-12-ATREG)に基づいてラベルに被験者ID、製造番号、使用期限を入力して印刷し、容器にラベルを貼付する。
・適切な基準(代表的には、NUHCPC-PMF-ATREG14)に基づいて「用法・用量・効能または効果ならびに使用上の注意または取扱い上の注意」を発行する。
・チャック付ビニール袋に試験物および「用法・用量・効能または効果ならびに使用上の注意または取扱い上の注意」を収納する。
・出荷するまで移送容器に入れてモニタリングユニット内に保管する。 8. Secondary Packaging In one embodiment, secondary packaging can be performed as follows: The various numerical values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can appropriately modify them to perform secondary packaging.
Typically, enter and print the subject ID, serial number, and expiration date on a label based on the appropriate standard (typically, NUHCPC-M-12-ATREG) and attach the label to the container.
- Issue "Dosage and administration, indications or effects, and precautions for use or handling" based on appropriate standards (typically, NUHCPC-PMF-ATREG14).
- Place the test item and "usage, dosage, efficacy or effects, and precautions for use or handling" in a plastic bag with a zipper.
- Store in a transport container in the monitoring unit until shipment.
一つの実施形態では、二次包装は以下のように行うことができる。以下に例示する各種数値、試薬や手順等は、代表例であり、当業者は適宜変更して二次包装を行うことができる。
・代表的には、適切な基準(代表的には、NUHCPC-M-12-ATREG)に基づいてラベルに被験者ID、製造番号、使用期限を入力して印刷し、容器にラベルを貼付する。
・適切な基準(代表的には、NUHCPC-PMF-ATREG14)に基づいて「用法・用量・効能または効果ならびに使用上の注意または取扱い上の注意」を発行する。
・チャック付ビニール袋に試験物および「用法・用量・効能または効果ならびに使用上の注意または取扱い上の注意」を収納する。
・出荷するまで移送容器に入れてモニタリングユニット内に保管する。 8. Secondary Packaging In one embodiment, secondary packaging can be performed as follows: The various numerical values, reagents, procedures, etc. exemplified below are representative examples, and those skilled in the art can appropriately modify them to perform secondary packaging.
Typically, enter and print the subject ID, serial number, and expiration date on a label based on the appropriate standard (typically, NUHCPC-M-12-ATREG) and attach the label to the container.
- Issue "Dosage and administration, indications or effects, and precautions for use or handling" based on appropriate standards (typically, NUHCPC-PMF-ATREG14).
- Place the test item and "usage, dosage, efficacy or effects, and precautions for use or handling" in a plastic bag with a zipper.
- Store in a transport container in the monitoring unit until shipment.
一つの実施形態では、細胞製剤は以下のように製造することができる。以下に例示する各種数値等は、代表例であり、当業者は適宜変更して細胞製剤の製造を行うことができる。
1) 投与19日前頃に、医療機関においてドナーにアフェレーシスを実施し、ドナーアフェレーシス産物を30Gyの放射線を照射し、細胞増殖能を無くした後に、細胞加工を実施する細胞培養加工施設へ発送する。
2) ドナーアフェレーシス産物受領後、細胞培養加工施設において、ドナー単核球を密度勾配遠心法により分離、回収後、2つに分けて凍結し、-80±10℃で保管する。
3) 投与14日前頃に、医療機関においてレシピエントにアフェレーシスを実施し、レシピエントアフェレーシス産物を、細胞加工を実施する細胞培養加工施設へ発送する。
4) レシピエントアフェレーシス産物受領後、細胞培養加工施設において、レシピエント単核球を密度勾配遠心法により分離、回収し、解凍したドナー単核球ならびに抗CD80抗体および抗CD86抗体またはCTLA4-Ig融合タンパク質などの阻害因子と共培養する。
5) 投与7日前頃に培地交換を行う。7日間培養した中間製品を回収し、解凍したドナー単核球ならびに抗CD80抗体および抗CD86抗体またはCTLA4-Ig融合タンパク質などの阻害因子と共培養する。
6) 投与当日に細胞加工物を密度勾配遠心法により回収後に、洗浄し、生理食塩液に充填する。
7) 医療機関に発送し、医療機関でレシピエントに投与する。 In one embodiment, the cell preparation can be produced as follows: The various numerical values exemplified below are representative examples, and a person skilled in the art can appropriately modify them to produce the cell preparation.
1) Approximately 19 days prior to administration, apheresis will be performed on the donor at a medical institution, and the donor apheresis product will be irradiated with 30 Gy of radiation to eliminate cell proliferation ability, and then shipped to a cell processing facility where cell processing will be performed.
2) After receiving the donor apheresis product, the donor mononuclear cells will be separated and collected by density gradient centrifugation at the cell culture processing facility, then divided into two aliquots, frozen, and stored at -80±10°C.
3) Approximately 14 days before administration, apheresis is performed on the recipient at a medical institution, and the recipient apheresis product is shipped to a cell culture processing facility where cell processing will be performed.
4) After receiving the recipient apheresis product, in a cell culture processing facility, the recipient mononuclear cells are separated and collected by density gradient centrifugation and co-cultured with thawed donor mononuclear cells and inhibitors such as anti-CD80 and anti-CD86 antibodies or CTLA4-Ig fusion protein.
5) The medium is replaced about 7 days before administration. The intermediate product cultured for 7 days is collected and co-cultured with thawed donor mononuclear cells and inhibitors such as anti-CD80 and anti-CD86 antibodies or CTLA4-Ig fusion protein.
6) On the day of administration, the processed cells are collected by density gradient centrifugation, washed, and filled with physiological saline.
7) Ship it to a medical institution and administer it to the recipient at the medical institution.
1) 投与19日前頃に、医療機関においてドナーにアフェレーシスを実施し、ドナーアフェレーシス産物を30Gyの放射線を照射し、細胞増殖能を無くした後に、細胞加工を実施する細胞培養加工施設へ発送する。
2) ドナーアフェレーシス産物受領後、細胞培養加工施設において、ドナー単核球を密度勾配遠心法により分離、回収後、2つに分けて凍結し、-80±10℃で保管する。
3) 投与14日前頃に、医療機関においてレシピエントにアフェレーシスを実施し、レシピエントアフェレーシス産物を、細胞加工を実施する細胞培養加工施設へ発送する。
4) レシピエントアフェレーシス産物受領後、細胞培養加工施設において、レシピエント単核球を密度勾配遠心法により分離、回収し、解凍したドナー単核球ならびに抗CD80抗体および抗CD86抗体またはCTLA4-Ig融合タンパク質などの阻害因子と共培養する。
5) 投与7日前頃に培地交換を行う。7日間培養した中間製品を回収し、解凍したドナー単核球ならびに抗CD80抗体および抗CD86抗体またはCTLA4-Ig融合タンパク質などの阻害因子と共培養する。
6) 投与当日に細胞加工物を密度勾配遠心法により回収後に、洗浄し、生理食塩液に充填する。
7) 医療機関に発送し、医療機関でレシピエントに投与する。 In one embodiment, the cell preparation can be produced as follows: The various numerical values exemplified below are representative examples, and a person skilled in the art can appropriately modify them to produce the cell preparation.
1) Approximately 19 days prior to administration, apheresis will be performed on the donor at a medical institution, and the donor apheresis product will be irradiated with 30 Gy of radiation to eliminate cell proliferation ability, and then shipped to a cell processing facility where cell processing will be performed.
2) After receiving the donor apheresis product, the donor mononuclear cells will be separated and collected by density gradient centrifugation at the cell culture processing facility, then divided into two aliquots, frozen, and stored at -80±10°C.
3) Approximately 14 days before administration, apheresis is performed on the recipient at a medical institution, and the recipient apheresis product is shipped to a cell culture processing facility where cell processing will be performed.
4) After receiving the recipient apheresis product, in a cell culture processing facility, the recipient mononuclear cells are separated and collected by density gradient centrifugation and co-cultured with thawed donor mononuclear cells and inhibitors such as anti-CD80 and anti-CD86 antibodies or CTLA4-Ig fusion protein.
5) The medium is replaced about 7 days before administration. The intermediate product cultured for 7 days is collected and co-cultured with thawed donor mononuclear cells and inhibitors such as anti-CD80 and anti-CD86 antibodies or CTLA4-Ig fusion protein.
6) On the day of administration, the processed cells are collected by density gradient centrifugation, washed, and filled with physiological saline.
7) Ship it to a medical institution and administer it to the recipient at the medical institution.
さらなる態様において、本開示は、免疫応答により媒介される被験体の疾患を処置するための組成物であって、該組成物が、IL-2とIL-2受容体(IL-2R)との相互作用を阻害する阻害因子および/またはCD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合することによって選択的に誘導された抗原特異的抑制性T細胞を含む、組成物を提供する。
In a further aspect, the disclosure provides a composition for treating a disease in a subject mediated by an immune response, the composition comprising an inhibitor that inhibits the interaction of IL-2 with the IL-2 receptor (IL-2R) and/or an inhibitor that can inhibit the interaction of CD80 and/or CD86 with CD28, and antigen-specific suppressor T cells selectively induced by mixing cells derived from the subject with an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
いくつかの実施形態において、疾患は、移植免疫拒絶反応、アレルギー、自己免疫疾患、移植片対宿主病、およびiPS細胞もしくはES細胞またはそれらから分化した細胞、組織もしくは臓器の移植によって引き起こされる免疫拒絶反応からなる群より選択され得る。
In some embodiments, the disease may be selected from the group consisting of transplant immune rejection, allergy, autoimmune disease, graft-versus-host disease, and immune rejection caused by transplantation of iPS cells or ES cells or cells, tissues, or organs differentiated therefrom.
いくつかの実施形態において、移植免疫拒絶反応は、腎臓、肝臓、心臓、皮膚、肺、すい臓、食道、胃、小腸、大腸、神経、血液、免疫系細胞を含む血球細胞、骨、軟骨、血管、角膜、眼球または骨髄が移植されることにより生じることを特徴とする。
In some embodiments, the transplant immune rejection is characterized as occurring following transplantation of kidney, liver, heart, skin, lung, pancreas, esophagus, stomach, small intestine, large intestine, nerves, blood, blood cells including immune system cells, bone, cartilage, blood vessels, cornea, eyeball, or bone marrow.
本開示が対象とする疾患等が移植免疫拒絶反応である実施形態において、アナジー細胞は、上記阻害因子と、レシピエント由来の細胞と、ドナー由来の抗原またはドナー由来の抗原の含有物とを混合することによって誘導され得る。ドナー由来の抗原の含有物は、PBMC、脾臓細胞または移植される臓器由来の細胞であり得る。
In an embodiment in which the disease or the like targeted by the present disclosure is transplant immune rejection, anergy cells can be induced by mixing the inhibitor, recipient-derived cells, and donor-derived antigens or donor-derived antigen-containing material. The donor-derived antigen-containing material can be PBMCs, spleen cells, or cells derived from the organ to be transplanted.
本開示が対象とする疾患等がアレルギーである実施形態において、アナジー細胞は、上記阻害因子と、被験体由来の細胞と、アレルギーを引き起こす被験体に由来しない抗原とを混合することによって誘導され得る。
In an embodiment in which the disease etc. targeted by the present disclosure is an allergy, anergy cells can be induced by mixing the inhibitor, cells derived from the subject, and an antigen not derived from the subject that causes the allergy.
本開示が対象とする疾患等が自己免疫疾患である実施形態において、アナジー細胞は、上記阻害因子と、被験体由来の細胞と、自己免疫疾患の原因となる被験体由来の抗原とを混合することによって誘導され得る。
In an embodiment in which the disease etc. targeted by the present disclosure is an autoimmune disease, anergy cells can be induced by mixing the inhibitor, cells derived from the subject, and an antigen derived from the subject that causes the autoimmune disease.
本開示が対象とする疾患等が移植片対宿主病である実施形態において、アナジー細胞は、上記阻害因子と、移植片を提供するドナーのPBMCまたは脾臓細胞と、レシピエント由来の抗原または該抗原の含有物とを混合することによって誘導され得る。レシピエント由来の抗原の含有物は、PBMC、脾臓細胞または臓器が移植される部位の周辺の細胞もしくはそれに由来する細胞であり得る。
In an embodiment in which the disease etc. targeted by the present disclosure is graft-versus-host disease, anergy cells can be induced by mixing the inhibitor, PBMCs or spleen cells of the donor providing the graft, and a recipient-derived antigen or a substance containing the antigen. The recipient-derived antigen can be PBMCs, spleen cells, or cells in the vicinity of the site where the organ is transplanted or cells derived therefrom.
本開示が対象とする疾患等がiPS細胞またはES細胞およびそれらの細胞から分化した細胞、組織または臓器の移植によって引き起こされる免疫拒絶反応である実施形態において、アナジー細胞は、上記阻害因子と、被験体由来の細胞と、iPS細胞またはES細胞から分化させた移植に用いる細胞とを混合することによって誘導され得る。
In an embodiment in which the disease etc. targeted by this disclosure is an immune rejection reaction caused by transplantation of iPS cells or ES cells and cells, tissues or organs differentiated from these cells, anergy cells can be induced by mixing the above-mentioned inhibitor with cells derived from the subject and cells differentiated from iPS cells or ES cells to be used for transplantation.
以下に本開示による疾患等の治療例を示すが、以下に限定されるものではない。
The following are examples of treatments for diseases and the like using this disclosure, but they are not limited to these.
(アレルギーおよび自己免疫疾患)
1つの局面において、本開示は、本開示の医薬を用いてアレルギーおよび/または自己免疫疾患を治療または予防する方法およびそれに用いる医薬、組成物、細胞混合物を提供する。アレルギーおよび自己免疫疾患に関しては、患者の末梢血から得たマクロファージを慣例的な方法により抗原提示能の高い樹状細胞(マクロファージ由来樹状細胞)へと分化させ、放射線(γ線)照射後のこの細胞にアレルギーや自己免疫疾患における過剰反応の原因となっている抗原を提示させ、同じ患者末梢血から得たT細胞群と、上記因子の存在下で1~2週間共培養し、アレルギーや自己免疫疾患の原因となっている抗原に特異的なアナジー細胞を得る。このアナジー細胞を患者に投与することで、アレルギーや自己免疫疾患の原因となっている抗原に特異的な免疫寛容を誘導し、アレルギーおよび自己免疫疾患の予防および治療に用いる。予防的療法であるか治療であるか、さらに症状の強弱等の諸条件により、投与回数は複数回となることもある。 (Allergies and autoimmune diseases)
In one aspect, the present disclosure provides a method for treating or preventing allergies and/or autoimmune diseases using the pharmaceutical of the present disclosure, and a pharmaceutical, composition, and cell mixture for use therein. Regarding allergies and autoimmune diseases, macrophages obtained from the peripheral blood of a patient are differentiated into dendritic cells (macrophage-derived dendritic cells) with high antigen-presenting ability by a conventional method, and these cells are made to present an antigen causing an overreaction in allergies or autoimmune diseases after irradiation (gamma rays), and co-cultured with a group of T cells obtained from the peripheral blood of the same patient in the presence of the above factors for 1 to 2 weeks to obtain anergy cells specific to the antigen causing the allergy or autoimmune disease. By administering these anergy cells to a patient, immune tolerance specific to the antigen causing the allergy or autoimmune disease is induced, and the anergy cells are used for the prevention and treatment of allergies and autoimmune diseases. The number of administrations may be multiple times depending on various conditions such as whether it is a preventive therapy or a treatment, and the strength of the symptoms.
1つの局面において、本開示は、本開示の医薬を用いてアレルギーおよび/または自己免疫疾患を治療または予防する方法およびそれに用いる医薬、組成物、細胞混合物を提供する。アレルギーおよび自己免疫疾患に関しては、患者の末梢血から得たマクロファージを慣例的な方法により抗原提示能の高い樹状細胞(マクロファージ由来樹状細胞)へと分化させ、放射線(γ線)照射後のこの細胞にアレルギーや自己免疫疾患における過剰反応の原因となっている抗原を提示させ、同じ患者末梢血から得たT細胞群と、上記因子の存在下で1~2週間共培養し、アレルギーや自己免疫疾患の原因となっている抗原に特異的なアナジー細胞を得る。このアナジー細胞を患者に投与することで、アレルギーや自己免疫疾患の原因となっている抗原に特異的な免疫寛容を誘導し、アレルギーおよび自己免疫疾患の予防および治療に用いる。予防的療法であるか治療であるか、さらに症状の強弱等の諸条件により、投与回数は複数回となることもある。 (Allergies and autoimmune diseases)
In one aspect, the present disclosure provides a method for treating or preventing allergies and/or autoimmune diseases using the pharmaceutical of the present disclosure, and a pharmaceutical, composition, and cell mixture for use therein. Regarding allergies and autoimmune diseases, macrophages obtained from the peripheral blood of a patient are differentiated into dendritic cells (macrophage-derived dendritic cells) with high antigen-presenting ability by a conventional method, and these cells are made to present an antigen causing an overreaction in allergies or autoimmune diseases after irradiation (gamma rays), and co-cultured with a group of T cells obtained from the peripheral blood of the same patient in the presence of the above factors for 1 to 2 weeks to obtain anergy cells specific to the antigen causing the allergy or autoimmune disease. By administering these anergy cells to a patient, immune tolerance specific to the antigen causing the allergy or autoimmune disease is induced, and the anergy cells are used for the prevention and treatment of allergies and autoimmune diseases. The number of administrations may be multiple times depending on various conditions such as whether it is a preventive therapy or a treatment, and the strength of the symptoms.
(移植片対宿主病)
1つの局面において、本開示は、本開示の医薬を用いて移植片対宿主病を治療または予防する方法およびそれに用いる医薬、組成物、細胞混合物を提供する。移植片対宿主病においては、移植免疫拒絶反応に対する治療とは対照的に、移植片を提供するドナーのPBMCまたはT細胞等の移植片対宿主病の原因となりうる細胞を、放射線(γ線)照射した宿主由来のPBMCまたはそれ以外の細胞と上記因子の存在下で1~2週間共培養し、宿主に特異的なアナジー細胞を得る。このアナジー細胞を宿主に投与することで、移植片対宿主病の原因となっている移植片による宿主への反応を抑制し(免疫寛容を誘導し)移植片対宿主病を予防および治療する。予防的療法であるか治療であるか、さらに移植する組織やその大きさ、症状の強弱等の諸条件により、投与回数は複数回となることもある。 (Graft-Versus-Host Disease)
In one aspect, the present disclosure provides a method for treating or preventing graft-versus-host disease using the medicament of the present disclosure, and a medicament, composition, and cell mixture for use therein. In contrast to treatment of transplant immune rejection, in graft-versus-host disease, cells that can cause graft-versus-host disease, such as PBMCs or T cells of a donor providing a graft, are co-cultured with PBMCs or other cells derived from a host irradiated with radiation (gamma rays) for 1 to 2 weeks in the presence of the above factors to obtain anergy cells specific to the host. By administering these anergy cells to a host, the reaction of the graft causing graft-versus-host disease to the host is suppressed (immune tolerance is induced), and graft-versus-host disease is prevented and treated. The number of administrations may be multiple, depending on various conditions, such as whether it is a preventive therapy or a treatment, the tissue to be transplanted, its size, and the severity of symptoms.
1つの局面において、本開示は、本開示の医薬を用いて移植片対宿主病を治療または予防する方法およびそれに用いる医薬、組成物、細胞混合物を提供する。移植片対宿主病においては、移植免疫拒絶反応に対する治療とは対照的に、移植片を提供するドナーのPBMCまたはT細胞等の移植片対宿主病の原因となりうる細胞を、放射線(γ線)照射した宿主由来のPBMCまたはそれ以外の細胞と上記因子の存在下で1~2週間共培養し、宿主に特異的なアナジー細胞を得る。このアナジー細胞を宿主に投与することで、移植片対宿主病の原因となっている移植片による宿主への反応を抑制し(免疫寛容を誘導し)移植片対宿主病を予防および治療する。予防的療法であるか治療であるか、さらに移植する組織やその大きさ、症状の強弱等の諸条件により、投与回数は複数回となることもある。 (Graft-Versus-Host Disease)
In one aspect, the present disclosure provides a method for treating or preventing graft-versus-host disease using the medicament of the present disclosure, and a medicament, composition, and cell mixture for use therein. In contrast to treatment of transplant immune rejection, in graft-versus-host disease, cells that can cause graft-versus-host disease, such as PBMCs or T cells of a donor providing a graft, are co-cultured with PBMCs or other cells derived from a host irradiated with radiation (gamma rays) for 1 to 2 weeks in the presence of the above factors to obtain anergy cells specific to the host. By administering these anergy cells to a host, the reaction of the graft causing graft-versus-host disease to the host is suppressed (immune tolerance is induced), and graft-versus-host disease is prevented and treated. The number of administrations may be multiple, depending on various conditions, such as whether it is a preventive therapy or a treatment, the tissue to be transplanted, its size, and the severity of symptoms.
(iPS細胞またはES細胞を用いた治療への応用)
1つの局面の局面において、本開示は、本開示の医薬を用いてiPS細胞またはES細胞を用いた予防または治療において、iPS細胞またはES細胞およびそれらの細胞から分化した細胞、組織または臓器の移植によって引き起こされる免疫拒絶反応またはその他の副作用を治療または予防する方法およびそれに用いる医薬、組成物、細胞混合物を提供する。iPS細胞またはES細胞を用いた治療への応用では、iPS細胞またはES細胞およびそれらの細胞から分化した細胞、組織または臓器の移植によって引き起こされる免疫拒絶反応が代表的に治療対象として例示される。 (Therapeutic application using iPS cells or ES cells)
In one aspect, the present disclosure provides a method for treating or preventing immune rejection or other side effects caused by transplantation of iPS cells or ES cells and cells, tissues or organs differentiated therefrom in prevention or therapy using iPS cells or ES cells using the pharmaceutical of the present disclosure, and a pharmaceutical, composition, and cell mixture used therein. In application to therapy using iPS cells or ES cells, immune rejection caused by transplantation of iPS cells or ES cells and cells, tissues or organs differentiated therefrom is typically exemplified as a treatment target.
1つの局面の局面において、本開示は、本開示の医薬を用いてiPS細胞またはES細胞を用いた予防または治療において、iPS細胞またはES細胞およびそれらの細胞から分化した細胞、組織または臓器の移植によって引き起こされる免疫拒絶反応またはその他の副作用を治療または予防する方法およびそれに用いる医薬、組成物、細胞混合物を提供する。iPS細胞またはES細胞を用いた治療への応用では、iPS細胞またはES細胞およびそれらの細胞から分化した細胞、組織または臓器の移植によって引き起こされる免疫拒絶反応が代表的に治療対象として例示される。 (Therapeutic application using iPS cells or ES cells)
In one aspect, the present disclosure provides a method for treating or preventing immune rejection or other side effects caused by transplantation of iPS cells or ES cells and cells, tissues or organs differentiated therefrom in prevention or therapy using iPS cells or ES cells using the pharmaceutical of the present disclosure, and a pharmaceutical, composition, and cell mixture used therein. In application to therapy using iPS cells or ES cells, immune rejection caused by transplantation of iPS cells or ES cells and cells, tissues or organs differentiated therefrom is typically exemplified as a treatment target.
代表的な例を詳細に述べると、iPS細胞またはES細胞を用いた治療への応用においては、iPS細胞またはES細胞から分化させた移植に用いる細胞または樹状細胞に放射線(γ線)を照射し、この細胞と移植を受ける患者のPBMCまたはT細胞群を上記因子の存在下で1~2週間共培養し、iPS細胞またはES細胞から分化させた細胞に特異的なアナジー細胞を得る。このアナジー細胞を宿主に投与することで、iPS細胞またはES細胞から分化した移植した細胞、組織、および臓器に特異的な免疫寛容を誘導し、それらへの拒絶反応を予防および治療する。予防的療法であるか治療であるか、さらに移植する組織やその大きさ、症状の強弱の諸条件により、投与回数は複数回となることもある。
To give a detailed description of a typical example, in therapeutic applications using iPS cells or ES cells, cells or dendritic cells to be used for transplantation that have been differentiated from iPS cells or ES cells are irradiated with radiation (gamma rays), and these cells are co-cultured with the PBMC or T cell group of the patient receiving the transplant in the presence of the above factors for 1-2 weeks to obtain anergy cells specific to the cells differentiated from iPS cells or ES cells. By administering these anergy cells to the host, specific immune tolerance is induced to the transplanted cells, tissues, and organs that have been differentiated from iPS cells or ES cells, and rejection reactions to them are prevented and treated. The number of administrations may be multiple depending on whether it is a preventive or therapeutic therapy, and on various conditions such as the tissue to be transplanted, its size, and the severity of symptoms.
(アレルギー)
1つの局面において、本開示は、本開示の医薬を用いてアレルギーを治療または予防する方法およびそれに用いる医薬、組成物、細胞混合物を提供する。本発明者らは、上記因子により抗原特異的にアナジーが誘導された細胞を含む製剤が、アレルギーに対する免疫寛容を惹起することができることを実証した。したがって、別の局面の局面において、本開示は、被験体のアレルギーを治療または予防するための組成物であって、該組成物は、上記因子と、該被験体由来の細胞と、アレルギーの原因となっている抗原または該抗原の含有物とを混合することによって免疫寛容が誘導された細胞を含む、組成物を提供する。 (allergy)
In one aspect, the present disclosure provides a method for treating or preventing allergy using the pharmaceutical of the present disclosure, and a pharmaceutical, composition, and cell mixture for use therein. The present inventors have demonstrated that a preparation containing cells in which anergy is induced antigen-specifically by the above-mentioned factor can induce immune tolerance to allergy. Therefore, in another aspect, the present disclosure provides a composition for treating or preventing allergy in a subject, the composition comprising cells in which immune tolerance is induced by mixing the above-mentioned factor, cells derived from the subject, and an antigen causing allergy or a substance containing the antigen.
1つの局面において、本開示は、本開示の医薬を用いてアレルギーを治療または予防する方法およびそれに用いる医薬、組成物、細胞混合物を提供する。本発明者らは、上記因子により抗原特異的にアナジーが誘導された細胞を含む製剤が、アレルギーに対する免疫寛容を惹起することができることを実証した。したがって、別の局面の局面において、本開示は、被験体のアレルギーを治療または予防するための組成物であって、該組成物は、上記因子と、該被験体由来の細胞と、アレルギーの原因となっている抗原または該抗原の含有物とを混合することによって免疫寛容が誘導された細胞を含む、組成物を提供する。 (allergy)
In one aspect, the present disclosure provides a method for treating or preventing allergy using the pharmaceutical of the present disclosure, and a pharmaceutical, composition, and cell mixture for use therein. The present inventors have demonstrated that a preparation containing cells in which anergy is induced antigen-specifically by the above-mentioned factor can induce immune tolerance to allergy. Therefore, in another aspect, the present disclosure provides a composition for treating or preventing allergy in a subject, the composition comprising cells in which immune tolerance is induced by mixing the above-mentioned factor, cells derived from the subject, and an antigen causing allergy or a substance containing the antigen.
アレルギーの原因となる抗原としては、食品、花粉、薬品、および金属などが挙げられ、より具体的には、ダニ抗原、卵白抗原、ミルク抗原、小麦抗原、ピーナッツ抗原、大豆抗原、ソバ抗原、ゴマ抗原、コメ抗原、甲殻類抗原、キウイ抗原、リンゴ抗原、バナナ抗原、モモ抗原、トマト抗原、マグロ抗原、サケ抗原、サバ抗原、牛肉抗原、鶏肉抗原、豚肉抗原、ネコ皮屑抗原、昆虫抗原、花粉抗原、イヌ皮屑抗原、真菌抗原、細菌抗原、ラテックス、ハプテンおよび金属等が挙げられるがこれらに限定されない。
Allergens that cause allergies include foods, pollen, medicines, and metals, and more specifically include, but are not limited to, dust mite antigens, egg white antigens, milk antigens, wheat antigens, peanut antigens, soybean antigens, buckwheat antigens, sesame antigens, rice antigens, crustacean antigens, kiwi antigens, apple antigens, banana antigens, peach antigens, tomato antigens, tuna antigens, salmon antigens, mackerel antigens, beef antigens, chicken antigens, pork antigens, cat dander antigens, insect antigens, pollen antigens, dog dander antigens, fungal antigens, bacterial antigens, latex, haptens, and metals.
(iPS細胞等によって生じる免疫拒絶反応の抑制または予防)
1つの局面の局面において、本開示は、iPS細胞等によって生じる免疫拒絶反応の抑制または予防のための方法を提供し、その抑制または予防のための、医薬、組成物、細胞混合物を提供する。本発明者らは、上記因子によりアナジーが誘導された細胞を含む製剤が、iPS細胞等またはそれらから分化した細胞、組織または臓器によって生じる免疫拒絶反応に対する免疫寛容を惹起することができることを実証した。したがって、別の局面の局面において、本開示は、被験体において、iPS細胞またはES細胞またはそれらから分化した細胞、組織もしくは臓器によって生じる免疫拒絶反応を抑制するまたは予防するための組成物であって、該組成物は、上記因子と、該被験体由来の細胞と、該iPS細胞またはES細胞から分化した抗原または該抗原の含有物とを混合することによって免疫寛容が誘導された細胞を含む、組成物を提供する。 (Suppression or prevention of immune rejection caused by iPS cells, etc.)
In one aspect, the present disclosure provides a method for suppressing or preventing immune rejection caused by iPS cells, etc., and provides a medicine, composition, and cell mixture for the suppression or prevention. The present inventors have demonstrated that a preparation containing cells in which anergy is induced by the above-mentioned factor can induce immune tolerance to immune rejection caused by iPS cells, etc., or cells, tissues, or organs differentiated therefrom. Therefore, in another aspect, the present disclosure provides a composition for suppressing or preventing immune rejection caused by iPS cells or ES cells, or cells, tissues, or organs differentiated therefrom, in a subject, the composition comprising cells in which immune tolerance has been induced by mixing the above-mentioned factor, cells derived from the subject, and an antigen differentiated from the iPS cells or ES cells, or a substance containing the antigen.
1つの局面の局面において、本開示は、iPS細胞等によって生じる免疫拒絶反応の抑制または予防のための方法を提供し、その抑制または予防のための、医薬、組成物、細胞混合物を提供する。本発明者らは、上記因子によりアナジーが誘導された細胞を含む製剤が、iPS細胞等またはそれらから分化した細胞、組織または臓器によって生じる免疫拒絶反応に対する免疫寛容を惹起することができることを実証した。したがって、別の局面の局面において、本開示は、被験体において、iPS細胞またはES細胞またはそれらから分化した細胞、組織もしくは臓器によって生じる免疫拒絶反応を抑制するまたは予防するための組成物であって、該組成物は、上記因子と、該被験体由来の細胞と、該iPS細胞またはES細胞から分化した抗原または該抗原の含有物とを混合することによって免疫寛容が誘導された細胞を含む、組成物を提供する。 (Suppression or prevention of immune rejection caused by iPS cells, etc.)
In one aspect, the present disclosure provides a method for suppressing or preventing immune rejection caused by iPS cells, etc., and provides a medicine, composition, and cell mixture for the suppression or prevention. The present inventors have demonstrated that a preparation containing cells in which anergy is induced by the above-mentioned factor can induce immune tolerance to immune rejection caused by iPS cells, etc., or cells, tissues, or organs differentiated therefrom. Therefore, in another aspect, the present disclosure provides a composition for suppressing or preventing immune rejection caused by iPS cells or ES cells, or cells, tissues, or organs differentiated therefrom, in a subject, the composition comprising cells in which immune tolerance has been induced by mixing the above-mentioned factor, cells derived from the subject, and an antigen differentiated from the iPS cells or ES cells, or a substance containing the antigen.
iPS細胞またはES細胞から分化した細胞、組織もしくは臓器としては、例えば、神経細胞または組織、角膜細胞または組織、心筋細胞または組織、肝臓または組織、軟骨細胞または組織、皮膚細胞または組織、腎臓または組織、などが挙げられるが、これらに限定されない。好ましい実施形態において、iPS細胞またはES細胞から分化した細胞、組織もしくは臓器としては、神経細胞または組織、心筋細胞または組織、軟骨細胞または組織および皮膚細胞または組織が挙げられる。
Cells, tissues, or organs differentiated from iPS cells or ES cells include, but are not limited to, nerve cells or tissue, corneal cells or tissue, cardiac muscle cells or tissue, liver or tissue, cartilage cells or tissue, skin cells or tissue, kidney or tissue, etc. In a preferred embodiment, cells, tissues, or organs differentiated from iPS cells or ES cells include nerve cells or tissue, cardiac muscle cells or tissue, cartilage cells or tissue, and skin cells or tissue.
(注記)
本明細書において「または」は、文章中に列挙されている事項の「少なくとも1つ以上」を採用できるときに使用される。「もしくは」も同様である。本明細書において「2つの値の範囲内」と明記した場合、その範囲には2つの値自体も含む。 (Note)
In this specification, "or" is used when "at least one or more" of the items listed in the sentence can be employed. The same applies to "alternative." In this specification, when it is specified that "within a range of two values," the range includes the two values themselves.
本明細書において「または」は、文章中に列挙されている事項の「少なくとも1つ以上」を採用できるときに使用される。「もしくは」も同様である。本明細書において「2つの値の範囲内」と明記した場合、その範囲には2つの値自体も含む。 (Note)
In this specification, "or" is used when "at least one or more" of the items listed in the sentence can be employed. The same applies to "alternative." In this specification, when it is specified that "within a range of two values," the range includes the two values themselves.
本明細書において引用された、科学文献、特許、特許出願などの参考文献は、その全体が、各々具体的に記載されたのと同じ程度に本明細書において参考として援用される。
All references cited herein, including scientific literature, patents, patent applications, and the like, are hereby incorporated by reference in their entirety to the same extent as if each was specifically set forth herein.
以上、本開示を、理解の容易のために好ましい実施形態を示して説明してきた。以下に、実施例に基づいて本開示を説明するが、上述の説明および以下の実施例は、例示の目的のみに提供され、本開示を限定する目的で提供したのではない。従って、本開示の範囲は、本明細書に具体的に記載された実施形態にも実施例にも限定されず、請求の範囲によってのみ限定される。
The present disclosure has been described above by showing preferred embodiments for ease of understanding. Below, the present disclosure will be described based on examples, but the above description and the following examples are provided for illustrative purposes only and are not provided for the purpose of limiting the present disclosure. Therefore, the scope of the present disclosure is not limited to the embodiments or examples specifically described in this specification, but is limited only by the scope of the claims.
以下、実施例に基づいて本開示をより具体的に説明する。ただし、本開示はこれらの実施例に限定されるものではない。なお、本明細書全体を通して引用される全文献は参照によりそのまま本願に組み込まれる。
The present disclosure will be described in more detail below with reference to examples. However, the present disclosure is not limited to these examples. All references cited throughout this specification are incorporated herein by reference in their entirety.
(抗体)
以下の実施例において使用した抗体は以下のとおりである。抗CD80モノクローナル抗体(mAb):クローンRM80抗CD86モノクローナル抗体(mAb):クローンGL-1抗IL-2モノクローナル抗体(mAb):InVivoMAb anti-mouse IL-2;メーカー:Bio X Cell;型番:BE0043(実施例1:IL-2添加によるTregのアロ特異的抑制への影響) 本実施例では、混合リンパ球反応(MLR)において、抗CD80/86抗体存在下でIL-2を添加すると、Tregの抗原特異的抑制能が減弱することを確認した。実施例1の概要を図1および3に示す。 (antibody)
The antibodies used in the following examples are as follows: Anti-CD80 monoclonal antibody (mAb): clone RM80 Anti-CD86 monoclonal antibody (mAb): clone GL-1 Anti-IL-2 monoclonal antibody (mAb): InVivoMAb anti-mouse IL-2; manufacturer: Bio X Cell; model number: BE0043 (Example 1: Effect of addition of IL-2 on allospecific suppression by Treg) In this example, it was confirmed that the addition of IL-2 in the presence of anti-CD80/86 antibodies in a mixed lymphocyte reaction (MLR) attenuates the antigen-specific suppressive ability of Treg. An overview of Example 1 is shown in Figures 1 and 3.
以下の実施例において使用した抗体は以下のとおりである。抗CD80モノクローナル抗体(mAb):クローンRM80抗CD86モノクローナル抗体(mAb):クローンGL-1抗IL-2モノクローナル抗体(mAb):InVivoMAb anti-mouse IL-2;メーカー:Bio X Cell;型番:BE0043(実施例1:IL-2添加によるTregのアロ特異的抑制への影響) 本実施例では、混合リンパ球反応(MLR)において、抗CD80/86抗体存在下でIL-2を添加すると、Tregの抗原特異的抑制能が減弱することを確認した。実施例1の概要を図1および3に示す。 (antibody)
The antibodies used in the following examples are as follows: Anti-CD80 monoclonal antibody (mAb): clone RM80 Anti-CD86 monoclonal antibody (mAb): clone GL-1 Anti-IL-2 monoclonal antibody (mAb): InVivoMAb anti-mouse IL-2; manufacturer: Bio X Cell; model number: BE0043 (Example 1: Effect of addition of IL-2 on allospecific suppression by Treg) In this example, it was confirmed that the addition of IL-2 in the presence of anti-CD80/86 antibodies in a mixed lymphocyte reaction (MLR) attenuates the antigen-specific suppressive ability of Treg. An overview of Example 1 is shown in Figures 1 and 3.
(方法)
1.Foxp3/hCD2-KI C57BL6マウス(hCD2-KIマウス)から脾臓細胞を回収し4×106個/mLに調製した。 (method)
1. Spleen cells were collected from Foxp3/hCD2-KI C57BL6 mice (hCD2-KI mice) and adjusted to 4 x 10 6 cells/mL.
1.Foxp3/hCD2-KI C57BL6マウス(hCD2-KIマウス)から脾臓細胞を回収し4×106個/mLに調製した。 (method)
1. Spleen cells were collected from Foxp3/hCD2-KI C57BL6 mice (hCD2-KI mice) and adjusted to 4 x 10 6 cells/mL.
2.Balb/cマウスの脾臓細胞を採取しX線30Gyを照射した後4×106個/mLの濃度に調製しhCD2-KIマウス脾臓細胞と1:1の比率で混合した。
2. Spleen cells were collected from Balb/c mice and irradiated with 30 Gy of X-rays, after which they were adjusted to a concentration of 4 x 106 cells/mL and mixed with hCD2-KI mouse spleen cells at a 1:1 ratio.
3.抗CD80mAb+抗CD86mAb(最終濃度各10μg/mL)および必要に応じて組換えIL-2(最終濃度25U/mL)を添加し、10cmディッシュに播種して37℃、5%CO2インキュベーターで7日間培養した。
3. Anti-CD80 mAb + anti-CD86 mAb (final concentration: 10 μg/mL each) and, if necessary, recombinant IL-2 (final concentration: 25 U/mL) were added, seeded on a 10 cm dish, and cultured at 37° C. in a 5% CO 2 incubator for 7 days.
4.培養7日後に細胞を回収し、必要に応じて磁気ビーズを用いてTregを分離した。
4. After 7 days of culture, cells were harvested and Tregs were isolated using magnetic beads as needed.
5.B6マウス脾臓細胞、LPS刺激Balb/cマウス脾臓細胞またはLPS刺激CBA/Nslcマウス脾臓細胞、および3.の培養した全細胞(whole cell)または分離したTregを表1の濃度に調製し、U底96ウェルプレートに播種した。
5. B6 mouse spleen cells, LPS-stimulated Balb/c mouse spleen cells, or LPS-stimulated CBA/Nslc mouse spleen cells, and the cultured whole cells or isolated Tregs from 3. were prepared to the concentrations shown in Table 1 and seeded onto a U-bottom 96-well plate.
6.37℃、5%CO2インキュベーターで3日間培養した。
6. The cells were cultured at 37°C in a 5% CO2 incubator for 3 days.
7.培養3日目に培養上清を回収し、-20℃にて凍結保存した。
7. On the third day of culture, the culture supernatant was collected and frozen at -20°C.
8.回収した培養上清は、ELISAにてIFN-γを測定した。
8. The collected culture supernatant was used to measure IFN-γ by ELISA.
(CFSEによるTregの分裂評価)
1.Foxp3/hCD2-KI C57BL6マウス(hCD2-KIマウス)から脾臓細胞を回収し2×106個/mLに調製した。 (Assessment of Treg division by CFSE)
1. Spleen cells were collected from Foxp3/hCD2-KI C57BL6 mice (hCD2-KI mice) and adjusted to 2 x 106 cells/mL.
1.Foxp3/hCD2-KI C57BL6マウス(hCD2-KIマウス)から脾臓細胞を回収し2×106個/mLに調製した。 (Assessment of Treg division by CFSE)
1. Spleen cells were collected from Foxp3/hCD2-KI C57BL6 mice (hCD2-KI mice) and adjusted to 2 x 106 cells/mL.
2.Balb/cマウスの脾臓細胞を採取しX線30Gyを照射した後2×106個/mLの濃度に調製しhCD2-KIマウス脾臓細胞と1:1の比率で混合した。
2. Spleen cells were collected from Balb/c mice and irradiated with 30 Gy of X-rays, then adjusted to a concentration of 2 x 106 cells/mL and mixed with hCD2-KI mouse spleen cells at a 1:1 ratio.
3.抗CD80mAb+抗CD86mAb(最終濃度各10μg/mL)または抗IL-2mAb(最終濃度10μg/mL)、および必要に応じて組換えIL-2(最終濃度25U/mL)を添加し、U底96ウェルプレートに200μLずつ播種して37℃、5%CO2インキュベーターで4日間培養した。
3. Anti-CD80 mAb + anti-CD86 mAb (final concentration 10 μg/mL each) or anti-IL-2 mAb (final concentration 10 μg/mL), and recombinant IL-2 (final concentration 25 U/mL) were added as required, and 200 μL each was seeded into a U-bottom 96-well plate and cultured at 37°C in a 5% CO2 incubator for 4 days.
4.培養4日後に細胞を回収し、死細胞をzombie Violetで染色した後、PerCP/Cy5.5-CD3抗体、APC-hCD2抗体で染色し、CD3+hCD2+CFSE-細胞の割合をFACSLyricにて測定した。
4. After 4 days of culture, the cells were harvested and dead cells were stained with zombie violet, then stained with PerCP/Cy5.5-CD3 antibody and APC-hCD2 antibody, and the percentage of CD3+hCD2+CFSE- cells was measured using FACSLyric.
(結果)
結果を図2に示す。抗CD80mAb+抗CD86mAb存在下で培養した場合、Tregの増殖が抑制された。また、StimulatorとしてLPS-Balb/cおよびwhole cellを用いた場合、IFN-γの産生が抑制され、単離されたTregを用いた場合、おり強くIFN-γの産生が抑制された。したがって、抗CD80mAb+抗CD86mAbは、抗原特異的な誘導型抑制性T細胞の増殖を促進すると考えられる。 (result)
The results are shown in Figure 2. When cultured in the presence of anti-CD80 mAb + anti-CD86 mAb, Treg proliferation was suppressed. Furthermore, when LPS-Balb/c and whole cells were used as stimulators, IFN-γ production was suppressed, and when isolated Tregs were used, IFN-γ production was strongly suppressed. Therefore, it is considered that anti-CD80 mAb + anti-CD86 mAb promotes the proliferation of antigen-specific induced suppressor T cells.
結果を図2に示す。抗CD80mAb+抗CD86mAb存在下で培養した場合、Tregの増殖が抑制された。また、StimulatorとしてLPS-Balb/cおよびwhole cellを用いた場合、IFN-γの産生が抑制され、単離されたTregを用いた場合、おり強くIFN-γの産生が抑制された。したがって、抗CD80mAb+抗CD86mAbは、抗原特異的な誘導型抑制性T細胞の増殖を促進すると考えられる。 (result)
The results are shown in Figure 2. When cultured in the presence of anti-CD80 mAb + anti-CD86 mAb, Treg proliferation was suppressed. Furthermore, when LPS-Balb/c and whole cells were used as stimulators, IFN-γ production was suppressed, and when isolated Tregs were used, IFN-γ production was strongly suppressed. Therefore, it is considered that anti-CD80 mAb + anti-CD86 mAb promotes the proliferation of antigen-specific induced suppressor T cells.
他方で、IL-2がさらに添加された場合、IL-2が添加されなかった場合と比べて、Tregがより多く増殖した。また、StimulatorとしてLPS-Balb/c、ならびにIL-2存在下で培養されたwhole cellおよびTregを使用した場合、IFN-γの産生が上昇し、免疫応答抑制能が減弱された(図4)。したがって、IL-2は、抗原非特異的なTregの増殖も促進し、抗原特異的な抑制能が失われたと考えられる。stimulatorとしてLPS刺激CBA/Nslcマウス脾臓細胞を用いた場合は、いずれの群も免疫応答が確認された。
On the other hand, when IL-2 was further added, Tregs proliferated more than when IL-2 was not added. Furthermore, when LPS-Balb/c, and whole cells and Tregs cultured in the presence of IL-2 were used as stimulators, IFN-γ production increased and the ability to suppress immune responses was attenuated (Figure 4). Therefore, it is thought that IL-2 also promoted the proliferation of antigen-nonspecific Tregs, resulting in the loss of antigen-specific suppressive ability. When LPS-stimulated CBA/Nslc mouse spleen cells were used as stimulators, immune responses were confirmed in both groups.
(実施例2:抗IL-2抗体添加MLRにおけるTregのアロ特異的抑制能評価)
本実施例では、MLRにおいて、抗IL-2抗体を添加すると、抗CD80/86抗体を添加した時と同様にTregに抗原特異的抑制能が誘導されることを確認した。実施例2の概要を図5に示す。 (Example 2: Evaluation of allospecific suppressive ability of Treg in MLR with addition of anti-IL-2 antibody)
In this example, it was confirmed that the addition of anti-IL-2 antibody in MLR induces antigen-specific suppressive ability in Tregs, similar to the case of the addition of anti-CD80/86 antibody. An overview of Example 2 is shown in Figure 5.
本実施例では、MLRにおいて、抗IL-2抗体を添加すると、抗CD80/86抗体を添加した時と同様にTregに抗原特異的抑制能が誘導されることを確認した。実施例2の概要を図5に示す。 (Example 2: Evaluation of allospecific suppressive ability of Treg in MLR with addition of anti-IL-2 antibody)
In this example, it was confirmed that the addition of anti-IL-2 antibody in MLR induces antigen-specific suppressive ability in Tregs, similar to the case of the addition of anti-CD80/86 antibody. An overview of Example 2 is shown in Figure 5.
(方法)
1.Foxp3/hCD2-KI C57BL6マウス(hCD2-KIマウス)から脾臓細胞を回収し4×106個/mLに調製した。 (method)
1. Spleen cells were collected from Foxp3/hCD2-KI C57BL6 mice (hCD2-KI mice) and adjusted to 4 x 10 6 cells/mL.
1.Foxp3/hCD2-KI C57BL6マウス(hCD2-KIマウス)から脾臓細胞を回収し4×106個/mLに調製した。 (method)
1. Spleen cells were collected from Foxp3/hCD2-KI C57BL6 mice (hCD2-KI mice) and adjusted to 4 x 10 6 cells/mL.
2.Balb/cマウスの脾臓細胞を採取しX線30Gyを照射した後4×106個/mLの濃度に調製しhCD2-KIマウス脾臓細胞と1:1の比率で混合した。
2. Spleen cells were collected from Balb/c mice and irradiated with 30 Gy of X-rays, after which they were adjusted to a concentration of 4 x 106 cells/mL and mixed with hCD2-KI mouse spleen cells at a 1:1 ratio.
3.抗CD80mAb+抗CD86mAb(最終濃度各10μg/mL)または抗IL-2mAb(最終濃度10μg/mL)を添加し、U底96ウェルプレートに播種して37℃、5%CO2インキュベーターで7日間培養した。
3. Anti-CD80 mAb + anti-CD86 mAb (final concentration 10 μg/mL each) or anti-IL-2 mAb (final concentration 10 μg/mL) was added, seeded on a U-bottom 96-well plate, and cultured at 37° C. in a 5% CO 2 incubator for 7 days.
4.培養7日後に細胞を回収し、必要に応じて磁気ビーズを用いてTregを分離した。
4. After 7 days of culture, cells were harvested and Tregs were isolated using magnetic beads as needed.
5.B6マウス脾臓細胞、LPS刺激Balb/cマウス脾臓細胞またはLPS刺激CBA/Nslcマウス脾臓細胞、および3.の培養した全細胞(whole cell)または分離したTregを表2の濃度に調製し、U底96ウェルプレートに播種した。
5. B6 mouse spleen cells, LPS-stimulated Balb/c mouse spleen cells, or LPS-stimulated CBA/Nslc mouse spleen cells, and the cultured whole cells or isolated Tregs from 3. were prepared to the concentrations shown in Table 2 and seeded onto a U-bottom 96-well plate.
6.37℃、5%CO2インキュベーターで3日間培養した。
6. The cells were cultured at 37°C in a 5% CO2 incubator for 3 days.
7.培養3日目に培養上清を回収し、-20℃にて凍結保存した。
7. On the third day of culture, the culture supernatant was collected and frozen at -20°C.
8.回収した培養上清は、ELISAにてIFN-γを測定した。
8. The collected culture supernatant was used to measure IFN-γ by ELISA.
(結果)
結果を図6に示す。抗IL-2抗体を添加すると、whole cellsでは、抗CD80/86抗体添加時ほどではないが、抑制能が認められた。一方、Tregでは抗CD80/86抗体添加と同等の抗原特異的抑制能が誘導された。stimulatorとしてLPS刺激CBA/Nslcマウス脾臓細胞を用いた場合は、いずれの群も免疫応答が確認された。 (result)
The results are shown in Figure 6. When anti-IL-2 antibody was added, suppressive ability was observed in whole cells, although not as strong as when anti-CD80/86 antibody was added. On the other hand, antigen-specific suppressive ability equivalent to that induced by addition of anti-CD80/86 antibody was induced in Treg. When LPS-stimulated CBA/Nslc mouse spleen cells were used as the stimulator, immune responses were observed in all groups.
結果を図6に示す。抗IL-2抗体を添加すると、whole cellsでは、抗CD80/86抗体添加時ほどではないが、抑制能が認められた。一方、Tregでは抗CD80/86抗体添加と同等の抗原特異的抑制能が誘導された。stimulatorとしてLPS刺激CBA/Nslcマウス脾臓細胞を用いた場合は、いずれの群も免疫応答が確認された。 (result)
The results are shown in Figure 6. When anti-IL-2 antibody was added, suppressive ability was observed in whole cells, although not as strong as when anti-CD80/86 antibody was added. On the other hand, antigen-specific suppressive ability equivalent to that induced by addition of anti-CD80/86 antibody was induced in Treg. When LPS-stimulated CBA/Nslc mouse spleen cells were used as the stimulator, immune responses were observed in all groups.
(考察)
IL-2を用いてTregを増殖させる過程が、ほぼ全てのTreg療法に存在する。しかし、IL-2によって、ポリクローナルな(バルクの)Tregの増殖を誘導することとなり、効果を減弱している可能性がある(図7)。抗CD80/86抗体または抗IL-2抗体の添加は、MLR時において産生される増殖因子IL-2の産生量を抑制することにより、ポリクローナルな(バルクの)Tregの増殖を抑制する。一方で、ドナー反応性TCRからの刺激と少量のIL-2に依存して、ドナー反応性を持つTregが選択的に生き残る。その結果、抗CD80/86抗体または抗IL-2抗体を添加したMLRにより誘導された細胞では、whole cellsおよびTregが、ドナー特異的な抑制能を発揮する(図7および図8)。 (Discussion)
The process of expanding Tregs using IL-2 is present in almost all Treg therapies. However, IL-2 induces the proliferation of polyclonal (bulk) Tregs, which may weaken the effect (Figure 7). The addition of anti-CD80/86 antibodies or anti-IL-2 antibodies suppresses the proliferation of polyclonal (bulk) Tregs by suppressing the production of the growth factor IL-2 produced during MLR. On the other hand, donor-reactive Tregs selectively survive depending on stimulation from donor-reactive TCRs and a small amount of IL-2. As a result, in cells induced by MLR with the addition of anti-CD80/86 antibodies or anti-IL-2 antibodies, whole cells and Tregs exert donor-specific suppressive abilities (Figures 7 and 8).
IL-2を用いてTregを増殖させる過程が、ほぼ全てのTreg療法に存在する。しかし、IL-2によって、ポリクローナルな(バルクの)Tregの増殖を誘導することとなり、効果を減弱している可能性がある(図7)。抗CD80/86抗体または抗IL-2抗体の添加は、MLR時において産生される増殖因子IL-2の産生量を抑制することにより、ポリクローナルな(バルクの)Tregの増殖を抑制する。一方で、ドナー反応性TCRからの刺激と少量のIL-2に依存して、ドナー反応性を持つTregが選択的に生き残る。その結果、抗CD80/86抗体または抗IL-2抗体を添加したMLRにより誘導された細胞では、whole cellsおよびTregが、ドナー特異的な抑制能を発揮する(図7および図8)。 (Discussion)
The process of expanding Tregs using IL-2 is present in almost all Treg therapies. However, IL-2 induces the proliferation of polyclonal (bulk) Tregs, which may weaken the effect (Figure 7). The addition of anti-CD80/86 antibodies or anti-IL-2 antibodies suppresses the proliferation of polyclonal (bulk) Tregs by suppressing the production of the growth factor IL-2 produced during MLR. On the other hand, donor-reactive Tregs selectively survive depending on stimulation from donor-reactive TCRs and a small amount of IL-2. As a result, in cells induced by MLR with the addition of anti-CD80/86 antibodies or anti-IL-2 antibodies, whole cells and Tregs exert donor-specific suppressive abilities (Figures 7 and 8).
(実施例3:培養中におけるIL-2の影響)
本実施例では、抗CD80/86抗体および/または抗IL-2抗体を用いた抗原特異的誘導型抑制性T細胞の誘導におけるIL-2の影響について確認した。方法は、実施例1および2と同様に行った。培養1日目~5日目の培養液中のIL-2の濃度をELISAにより測定した。 Example 3: Effect of IL-2 during culture
In this example, the effect of IL-2 on the induction of antigen-specific inducible suppressor T cells using anti-CD80/86 antibody and/or anti-IL-2 antibody was confirmed. The method was the same as in Examples 1 and 2. The concentration of IL-2 in the culture medium on days 1 to 5 of culture was measured by ELISA.
本実施例では、抗CD80/86抗体および/または抗IL-2抗体を用いた抗原特異的誘導型抑制性T細胞の誘導におけるIL-2の影響について確認した。方法は、実施例1および2と同様に行った。培養1日目~5日目の培養液中のIL-2の濃度をELISAにより測定した。 Example 3: Effect of IL-2 during culture
In this example, the effect of IL-2 on the induction of antigen-specific inducible suppressor T cells using anti-CD80/86 antibody and/or anti-IL-2 antibody was confirmed. The method was the same as in Examples 1 and 2. The concentration of IL-2 in the culture medium on days 1 to 5 of culture was measured by ELISA.
(結果)
図9および図10は、抗CD80/86抗体または抗IL-2抗体の存在下で培養した場合は、IL-2濃度は、5日目において20pg/mL以下であったことを示す。抗CD80/86抗体によりCD28共刺激シグナルが阻害されるため、T細胞の活性化が低下しIL-2産生が抑制されたと考えられる。他方で、抗CD80/86抗体も抗IL-2抗体も用いずに培養した場合、IL-2は培養2日目においてすでに100pg/mLを超えるIL-2濃度を示した。 (result)
9 and 10 show that when cells were cultured in the presence of anti-CD80/86 antibody or anti-IL-2 antibody, the IL-2 concentration was 20 pg/mL or less on day 5. It is believed that the CD28 costimulatory signal was inhibited by anti-CD80/86 antibody, which reduced T cell activation and suppressed IL-2 production. On the other hand, when cells were cultured without anti-CD80/86 antibody or anti-IL-2 antibody, the IL-2 concentration exceeded 100 pg/mL already on day 2 of culture.
図9および図10は、抗CD80/86抗体または抗IL-2抗体の存在下で培養した場合は、IL-2濃度は、5日目において20pg/mL以下であったことを示す。抗CD80/86抗体によりCD28共刺激シグナルが阻害されるため、T細胞の活性化が低下しIL-2産生が抑制されたと考えられる。他方で、抗CD80/86抗体も抗IL-2抗体も用いずに培養した場合、IL-2は培養2日目においてすでに100pg/mLを超えるIL-2濃度を示した。 (result)
9 and 10 show that when cells were cultured in the presence of anti-CD80/86 antibody or anti-IL-2 antibody, the IL-2 concentration was 20 pg/mL or less on day 5. It is believed that the CD28 costimulatory signal was inhibited by anti-CD80/86 antibody, which reduced T cell activation and suppressed IL-2 production. On the other hand, when cells were cultured without anti-CD80/86 antibody or anti-IL-2 antibody, the IL-2 concentration exceeded 100 pg/mL already on day 2 of culture.
(実施例4:抗CD80/86抗体または抗IL-2抗体によるCD4+T細胞の割合の変化)
本実施例では、抗CD80/86抗体および/または抗IL-2抗体によるCD4+T細胞の割合の変化を確認した。方法は、実施例1および2と同様に行った。 (Example 4: Change in the percentage of CD4 + T cells by anti-CD80/86 antibody or anti-IL-2 antibody)
In this example, changes in the proportion of CD4 + T cells due to anti-CD80/86 antibody and/or anti-IL-2 antibody were confirmed. The methods were similar to those in Examples 1 and 2.
本実施例では、抗CD80/86抗体および/または抗IL-2抗体によるCD4+T細胞の割合の変化を確認した。方法は、実施例1および2と同様に行った。 (Example 4: Change in the percentage of CD4 + T cells by anti-CD80/86 antibody or anti-IL-2 antibody)
In this example, changes in the proportion of CD4 + T cells due to anti-CD80/86 antibody and/or anti-IL-2 antibody were confirmed. The methods were similar to those in Examples 1 and 2.
(結果)
結果を図11に示す。抗CD80/86抗体を添加した場合、「培養添加なし」と比較して、全細胞に対するCD4+T細胞およびTreg(CD4+FOXP3+T細胞)の割合を増加させ、CD4+T細胞に対するTregの割合も増加させた。抗IL-2中和抗体の添加の添加により、「培養添加なし」と比較して、全細胞に対するCD4+T細胞およびTreg(CD4+FOXP3+T細胞)の割合の増加は認められなかった。 (result)
The results are shown in Figure 11. When anti-CD80/86 antibody was added, the ratio of CD4 + T cells and Treg (CD4 + FOXP3 + T cells) to total cells was increased, and the ratio of Treg to CD4 + T cells was also increased, compared to "no culture addition." The addition of anti-IL-2 neutralizing antibody did not increase the ratio of CD4 + T cells and Treg (CD4 + FOXP3 + T cells) to total cells, compared to "no culture addition. "
結果を図11に示す。抗CD80/86抗体を添加した場合、「培養添加なし」と比較して、全細胞に対するCD4+T細胞およびTreg(CD4+FOXP3+T細胞)の割合を増加させ、CD4+T細胞に対するTregの割合も増加させた。抗IL-2中和抗体の添加の添加により、「培養添加なし」と比較して、全細胞に対するCD4+T細胞およびTreg(CD4+FOXP3+T細胞)の割合の増加は認められなかった。 (result)
The results are shown in Figure 11. When anti-CD80/86 antibody was added, the ratio of CD4 + T cells and Treg (CD4 + FOXP3 + T cells) to total cells was increased, and the ratio of Treg to CD4 + T cells was also increased, compared to "no culture addition." The addition of anti-IL-2 neutralizing antibody did not increase the ratio of CD4 + T cells and Treg (CD4 + FOXP3 + T cells) to total cells, compared to "no culture addition. "
(実施例5:抗CD80/86抗体または抗IL-2抗体による抑制能の変化)
本実施例では、抗CD80/86抗体および/または抗IL-2抗体による抑制能の変化を確認した。 (Example 5: Change in suppressive ability by anti-CD80/86 antibody or anti-IL-2 antibody)
In this example, changes in suppressive ability due to anti-CD80/86 antibody and/or anti-IL-2 antibody were confirmed.
本実施例では、抗CD80/86抗体および/または抗IL-2抗体による抑制能の変化を確認した。 (Example 5: Change in suppressive ability by anti-CD80/86 antibody or anti-IL-2 antibody)
In this example, changes in suppressive ability due to anti-CD80/86 antibody and/or anti-IL-2 antibody were confirmed.
(結果)
抗CD80/86抗体および抗IL-2抗体の両方を用いてIL-2を完全にブロックした場合は、Balb刺激に対して、単離したTregでは抑制能が認められるが、Whole cellでは抑制能がほとんど認められなかった。抗CD80/86抗体のみを添加して培養した場合は、Balb刺激に対して、より強い抑制能を示した。これらのことから、抑制性T細胞の抑制能はIL-2依存的であることが示された。単離したTregは、CBAに対してもわずかに抑制能を示したが、Whole cellではほとんど抑制能を示さなかった(図12および図13)。したがって、Whole cellの抑制能の抗原特性は高く、IL-2依存性も高い。抗原非特異的抑制は抑制性サイトカインTGF-β等の影響の可能性が考えられる。 (result)
When IL-2 was completely blocked using both anti-CD80/86 antibody and anti-IL-2 antibody, isolated Tregs exhibited suppressive ability against Balb stimulation, but whole cells exhibited almost no suppressive ability. When cultured with only anti-CD80/86 antibody, stronger suppressive ability against Balb stimulation was observed. These results indicate that the suppressive ability of suppressive T cells is IL-2 dependent. Isolated Tregs also exhibited slight suppressive ability against CBA, but whole cells exhibited almost no suppressive ability (FIGS. 12 and 13). Therefore, the suppressive ability of whole cells has high antigen specificity and is also highly dependent on IL-2. Antigen non-specific suppression may be due to the influence of the suppressive cytokine TGF-β, etc.
抗CD80/86抗体および抗IL-2抗体の両方を用いてIL-2を完全にブロックした場合は、Balb刺激に対して、単離したTregでは抑制能が認められるが、Whole cellでは抑制能がほとんど認められなかった。抗CD80/86抗体のみを添加して培養した場合は、Balb刺激に対して、より強い抑制能を示した。これらのことから、抑制性T細胞の抑制能はIL-2依存的であることが示された。単離したTregは、CBAに対してもわずかに抑制能を示したが、Whole cellではほとんど抑制能を示さなかった(図12および図13)。したがって、Whole cellの抑制能の抗原特性は高く、IL-2依存性も高い。抗原非特異的抑制は抑制性サイトカインTGF-β等の影響の可能性が考えられる。 (result)
When IL-2 was completely blocked using both anti-CD80/86 antibody and anti-IL-2 antibody, isolated Tregs exhibited suppressive ability against Balb stimulation, but whole cells exhibited almost no suppressive ability. When cultured with only anti-CD80/86 antibody, stronger suppressive ability against Balb stimulation was observed. These results indicate that the suppressive ability of suppressive T cells is IL-2 dependent. Isolated Tregs also exhibited slight suppressive ability against CBA, but whole cells exhibited almost no suppressive ability (FIGS. 12 and 13). Therefore, the suppressive ability of whole cells has high antigen specificity and is also highly dependent on IL-2. Antigen non-specific suppression may be due to the influence of the suppressive cytokine TGF-β, etc.
(実施例6:抗CD80/86抗体または抗IL-2中和抗体によるIL-2産生抑制の評価)
本実施例では、ヒト細胞を用いて、抗CD80/86抗体または抗IL-2中和抗体により、IL-2産生が抑制されることを確認する。 Example 6: Evaluation of inhibition of IL-2 production by anti-CD80/86 antibody or anti-IL-2 neutralizing antibody
In this example, using human cells, it is confirmed that IL-2 production is suppressed by anti-CD80/86 antibody or anti-IL-2 neutralizing antibody.
本実施例では、ヒト細胞を用いて、抗CD80/86抗体または抗IL-2中和抗体により、IL-2産生が抑制されることを確認する。 Example 6: Evaluation of inhibition of IL-2 production by anti-CD80/86 antibody or anti-IL-2 neutralizing antibody
In this example, using human cells, it is confirmed that IL-2 production is suppressed by anti-CD80/86 antibody or anti-IL-2 neutralizing antibody.
(材料)
・Responder-PBMC 4x106個/mL 100μL
・X線照射済みStimulator-PBMC 4x106個/mL 100μL
・抗CD80/86抗体 最終濃度10μg/mL
・抗IL-2中和抗体 最終濃度10μg/mL
(方法)
1.3人の健常人から末梢血リンパ球(PBMC)を採取する。
2.3通りのペアを組み、Stimulatorとなる細胞に30GyのX線を照射する。
3.以下の条件で96wellプレートに播種し、37℃,5%CO2条件下で培養する。
・Responder PBMC 4×106個/mL 100μL
・X線照射済みStimulator PBMC 4x106個/mL 100μL
・抗CD80/86抗体 最終濃度10μg/mL
・抗IL-2中和抗体 最終濃度10μg/mL
4.培養1,2,3,5日目に培養上清を回収し、ELISAによるIL-2測定日まで-20℃で凍結保存する。
5.ヒトIL-2のELISA測定をする。 (material)
・Responder-PBMC 4x10 6 pieces/mL 100μL
・X-ray irradiated Stimulator-PBMC 4x10 6 pieces/mL 100μL
・Anti-CD80/86 antibody final concentration 10μg/mL
・Anti-IL-2 neutralizing antibody final concentration 10μg/mL
(method)
1. Peripheral blood lymphocytes (PBMCs) are collected from three healthy individuals.
2. Three pairs were made and the stimulator cells were irradiated with 30 Gy of X-rays.
3. Seed the cells on a 96-well plate under the following conditions and culture them at 37°C and 5% CO2.
・Responder PBMC 4×10 6 pieces/mL 100 μL
・X-ray irradiated Stimulator PBMC 4x10 6 pieces/mL 100μL
・Anti-CD80/86 antibody final concentration 10μg/mL
・Anti-IL-2 neutralizing antibody final concentration 10μg/mL
4. The culture supernatant is collected on days 1, 2, 3, and 5 of culture, and frozen and stored at -20°C until the day of measurement of IL-2 by ELISA.
5. Measure human IL-2 by ELISA.
・Responder-PBMC 4x106個/mL 100μL
・X線照射済みStimulator-PBMC 4x106個/mL 100μL
・抗CD80/86抗体 最終濃度10μg/mL
・抗IL-2中和抗体 最終濃度10μg/mL
(方法)
1.3人の健常人から末梢血リンパ球(PBMC)を採取する。
2.3通りのペアを組み、Stimulatorとなる細胞に30GyのX線を照射する。
3.以下の条件で96wellプレートに播種し、37℃,5%CO2条件下で培養する。
・Responder PBMC 4×106個/mL 100μL
・X線照射済みStimulator PBMC 4x106個/mL 100μL
・抗CD80/86抗体 最終濃度10μg/mL
・抗IL-2中和抗体 最終濃度10μg/mL
4.培養1,2,3,5日目に培養上清を回収し、ELISAによるIL-2測定日まで-20℃で凍結保存する。
5.ヒトIL-2のELISA測定をする。 (material)
・Responder-PBMC 4x10 6 pieces/mL 100μL
・X-ray irradiated Stimulator-PBMC 4x10 6 pieces/mL 100μL
・Anti-CD80/86 antibody final concentration 10μg/mL
・Anti-IL-2 neutralizing antibody final concentration 10μg/mL
(method)
1. Peripheral blood lymphocytes (PBMCs) are collected from three healthy individuals.
2. Three pairs were made and the stimulator cells were irradiated with 30 Gy of X-rays.
3. Seed the cells on a 96-well plate under the following conditions and culture them at 37°C and 5% CO2.
・Responder PBMC 4×10 6 pieces/mL 100 μL
・X-ray irradiated Stimulator PBMC 4x10 6 pieces/mL 100μL
・Anti-CD80/86 antibody final concentration 10μg/mL
・Anti-IL-2 neutralizing antibody final concentration 10μg/mL
4. The culture supernatant is collected on days 1, 2, 3, and 5 of culture, and frozen and stored at -20°C until the day of measurement of IL-2 by ELISA.
5. Measure human IL-2 by ELISA.
(結果)
抗CD80/86抗体または抗IL-2抗体の存在下で培養した場合は、培養上清中のIL-2濃度は無添加群よりも低下すると予測される。 (result)
When cultured in the presence of anti-CD80/86 antibody or anti-IL-2 antibody, the IL-2 concentration in the culture supernatant is predicted to be lower than that in the control group.
抗CD80/86抗体または抗IL-2抗体の存在下で培養した場合は、培養上清中のIL-2濃度は無添加群よりも低下すると予測される。 (result)
When cultured in the presence of anti-CD80/86 antibody or anti-IL-2 antibody, the IL-2 concentration in the culture supernatant is predicted to be lower than that in the control group.
(実施例7:培養液中IL-2濃度の調節による、Tregのドナー特異的抑制能への影響の評価)
(方法)
1.Foxp3/hCD2-KI C57BL6マウス(hCD2-KIマウス)から脾臓細胞を回収し4×106個/mLに調製する。
2.Balb/cマウスの脾臓細胞を採取しX線30Gyを照射した後4×106個/mLの濃度に調製し、hCD2-KIマウス脾臓細胞と1:1の比率で混合する。
3.抗IL-2mAb(最終濃度10μg/mL)を添加し、10cmディッシュに播種して37℃、5%CO2インキュベーターで培養する。
4.培養2日目、4日目にIL-2を15pg/mLの濃度となるように添加する。
5.培養7日後に細胞を回収し、必要に応じて磁気ビーズを用いてTregを分離する。
6.B6マウス脾臓細胞、LPS刺激Balb/cマウス脾臓細胞またはLPS刺激CBA/Nslcマウス脾臓細胞、および5.の培養した全細胞(whole cell)または分離したTregを表1の濃度に調製し、U底96ウェルプレートに播種する。
7.37℃、5%CO2インキュベーターで3日間培養する。 (Example 7: Evaluation of the effect of adjusting the IL-2 concentration in the culture medium on the donor-specific suppressive ability of Treg)
(method)
1. Spleen cells are collected from Foxp3/hCD2-KI C57BL6 mice (hCD2-KI mice) and adjusted to 4 x 10 6 cells/mL.
2. Spleen cells from Balb/c mice are collected and irradiated with 30 Gy of X-rays, then adjusted to a concentration of 4 x 10 6 cells/mL and mixed with hCD2-KI mouse spleen cells at a 1:1 ratio.
3. Anti-IL-2 mAb (final concentration: 10 μg/mL) is added, the cells are seeded onto a 10 cm dish, and cultured at 37° C. in a 5% CO2 incubator.
4. On days 2 and 4 of culture, add IL-2 to a concentration of 15 pg/mL.
5. After 7 days of culture, the cells are harvested and, if necessary, Tregs are isolated using magnetic beads.
6. B6 mouse spleen cells, LPS-stimulated Balb/c mouse spleen cells or LPS-stimulated CBA/Nslc mouse spleen cells, and the cultured whole cells or isolated Tregs from 5. are prepared to the concentrations shown in Table 1 and seeded on a U-bottom 96-well plate.
7. Culture in a 37°C, 5% CO2 incubator for 3 days.
(方法)
1.Foxp3/hCD2-KI C57BL6マウス(hCD2-KIマウス)から脾臓細胞を回収し4×106個/mLに調製する。
2.Balb/cマウスの脾臓細胞を採取しX線30Gyを照射した後4×106個/mLの濃度に調製し、hCD2-KIマウス脾臓細胞と1:1の比率で混合する。
3.抗IL-2mAb(最終濃度10μg/mL)を添加し、10cmディッシュに播種して37℃、5%CO2インキュベーターで培養する。
4.培養2日目、4日目にIL-2を15pg/mLの濃度となるように添加する。
5.培養7日後に細胞を回収し、必要に応じて磁気ビーズを用いてTregを分離する。
6.B6マウス脾臓細胞、LPS刺激Balb/cマウス脾臓細胞またはLPS刺激CBA/Nslcマウス脾臓細胞、および5.の培養した全細胞(whole cell)または分離したTregを表1の濃度に調製し、U底96ウェルプレートに播種する。
7.37℃、5%CO2インキュベーターで3日間培養する。 (Example 7: Evaluation of the effect of adjusting the IL-2 concentration in the culture medium on the donor-specific suppressive ability of Treg)
(method)
1. Spleen cells are collected from Foxp3/hCD2-KI C57BL6 mice (hCD2-KI mice) and adjusted to 4 x 10 6 cells/mL.
2. Spleen cells from Balb/c mice are collected and irradiated with 30 Gy of X-rays, then adjusted to a concentration of 4 x 10 6 cells/mL and mixed with hCD2-KI mouse spleen cells at a 1:1 ratio.
3. Anti-IL-2 mAb (final concentration: 10 μg/mL) is added, the cells are seeded onto a 10 cm dish, and cultured at 37° C. in a 5% CO2 incubator.
4. On days 2 and 4 of culture, add IL-2 to a concentration of 15 pg/mL.
5. After 7 days of culture, the cells are harvested and, if necessary, Tregs are isolated using magnetic beads.
6. B6 mouse spleen cells, LPS-stimulated Balb/c mouse spleen cells or LPS-stimulated CBA/Nslc mouse spleen cells, and the cultured whole cells or isolated Tregs from 5. are prepared to the concentrations shown in Table 1 and seeded on a U-bottom 96-well plate.
7. Culture in a 37°C, 5% CO2 incubator for 3 days.
具体的には以下のように行った。
(IL-2測定)
2種類の市販ヒト末梢血単核細胞(LONZA社 CC-2702)をドナー細胞とレシピエント細胞として使用した。ドナー細胞(Batch#22TL290862)を15mLの10% FCS(SELBORNE社)含有RPMI培地(富士フィルム和光純薬)に懸濁後、25Gyの放射線を照射した。次に抗CD80抗体((株)JUNTEN BIO社製)をドナー細胞とレシピエント細胞(Batch#23TL043676)にそれぞれ最終濃度5 mg/Lで添加後、氷中に30分静置した。その後、遠心分離でドナー細胞とレシピエント細胞を回収し、ドナー細胞にCD86抗体((株)JUNTEN BIO社製)を最終濃度5 mg/Lで添加した。次に10% FCS含有RPMI培地を20 mL添加したTフラスコ(Thermo scientific社 25 cm2)2本にドナー細胞とレシピエント細胞がそれぞれ最終濃度 5.0x10^7 cells/20mLになるように調製した。Tフラスコ1本には、抗CD80抗体((株)JUNTEN BIO社製)と抗CD86抗体((株)JUNTEN BIO社製)を最終濃度5 mg/Lになるように添加し、残りの1本には抗体を添加せず陰性対照とした。2本のTフラスコは37℃に設定した5%CO2インキュベーター内に静置した。培養開始前、培養開始後1,2,3,4,7日目に培養上清を回収し、Cytometric Bead Array (CBA)法にてIL-2濃度を測定した結果を表3に示す。抗CD80と抗CD86抗体を添加した場合、陰性対照に比べて7日間の培養期間中4日目以降に著しくIL-2濃度が低下することが認められた。 Specifically, the following was carried out.
(IL-2 Measurement)
Two types of commercially available human peripheral blood mononuclear cells (LONZA CC-2702) were used as donor and recipient cells. The donor cells (Batch#22TL290862) were suspended in 15mL of RPMI medium (FUJIFILM Wako Pure Chemical Industries, Ltd.) containing 10% FCS (SELBORNE), and then irradiated with 25Gy of radiation. Next, anti-CD80 antibody (manufactured by JUNTEN BIO Co., Ltd.) was added to the donor cells and recipient cells (Batch#23TL043676) at a final concentration of 5 mg/L, respectively, and then allowed to stand on ice for 30 minutes. Thereafter, the donor cells and recipient cells were collected by centrifugation, and CD86 antibody (manufactured by JUNTEN BIO Co., Ltd.) was added to the donor cells at a final concentration of 5 mg/L. Next, donor cells and recipient cells were prepared to a final concentration of 5.0x10^7 cells/20mL in two T flasks (Thermo scientific, 25 cm2 ) to which 20mL of 10% FCS-containing RPMI medium had been added. Anti-CD80 antibody (manufactured by JUNTEN BIO Co., Ltd.) and anti-CD86 antibody (manufactured by JUNTEN BIO Co., Ltd.) were added to one T flask to a final concentration of 5mg/L, and the remaining one was used as a negative control without adding any antibody. The two T flasks were placed in a 5% CO2 incubator set at 37°C. The culture supernatant was collected before the start of the culture and on days 1, 2, 3, 4, and 7 after the start of the culture, and the IL-2 concentration was measured by the Cytometric Bead Array (CBA) method. The results are shown in Table 3. When anti-CD80 and anti-CD86 antibodies were added, the IL-2 concentration was found to be significantly reduced from day 4 onwards during the 7-day culture period, compared to the negative control.
(IL-2測定)
2種類の市販ヒト末梢血単核細胞(LONZA社 CC-2702)をドナー細胞とレシピエント細胞として使用した。ドナー細胞(Batch#22TL290862)を15mLの10% FCS(SELBORNE社)含有RPMI培地(富士フィルム和光純薬)に懸濁後、25Gyの放射線を照射した。次に抗CD80抗体((株)JUNTEN BIO社製)をドナー細胞とレシピエント細胞(Batch#23TL043676)にそれぞれ最終濃度5 mg/Lで添加後、氷中に30分静置した。その後、遠心分離でドナー細胞とレシピエント細胞を回収し、ドナー細胞にCD86抗体((株)JUNTEN BIO社製)を最終濃度5 mg/Lで添加した。次に10% FCS含有RPMI培地を20 mL添加したTフラスコ(Thermo scientific社 25 cm2)2本にドナー細胞とレシピエント細胞がそれぞれ最終濃度 5.0x10^7 cells/20mLになるように調製した。Tフラスコ1本には、抗CD80抗体((株)JUNTEN BIO社製)と抗CD86抗体((株)JUNTEN BIO社製)を最終濃度5 mg/Lになるように添加し、残りの1本には抗体を添加せず陰性対照とした。2本のTフラスコは37℃に設定した5%CO2インキュベーター内に静置した。培養開始前、培養開始後1,2,3,4,7日目に培養上清を回収し、Cytometric Bead Array (CBA)法にてIL-2濃度を測定した結果を表3に示す。抗CD80と抗CD86抗体を添加した場合、陰性対照に比べて7日間の培養期間中4日目以降に著しくIL-2濃度が低下することが認められた。 Specifically, the following was carried out.
(IL-2 Measurement)
Two types of commercially available human peripheral blood mononuclear cells (LONZA CC-2702) were used as donor and recipient cells. The donor cells (Batch#22TL290862) were suspended in 15mL of RPMI medium (FUJIFILM Wako Pure Chemical Industries, Ltd.) containing 10% FCS (SELBORNE), and then irradiated with 25Gy of radiation. Next, anti-CD80 antibody (manufactured by JUNTEN BIO Co., Ltd.) was added to the donor cells and recipient cells (Batch#23TL043676) at a final concentration of 5 mg/L, respectively, and then allowed to stand on ice for 30 minutes. Thereafter, the donor cells and recipient cells were collected by centrifugation, and CD86 antibody (manufactured by JUNTEN BIO Co., Ltd.) was added to the donor cells at a final concentration of 5 mg/L. Next, donor cells and recipient cells were prepared to a final concentration of 5.0x10^7 cells/20mL in two T flasks (Thermo scientific, 25 cm2 ) to which 20mL of 10% FCS-containing RPMI medium had been added. Anti-CD80 antibody (manufactured by JUNTEN BIO Co., Ltd.) and anti-CD86 antibody (manufactured by JUNTEN BIO Co., Ltd.) were added to one T flask to a final concentration of 5mg/L, and the remaining one was used as a negative control without adding any antibody. The two T flasks were placed in a 5% CO2 incubator set at 37°C. The culture supernatant was collected before the start of the culture and on days 1, 2, 3, 4, and 7 after the start of the culture, and the IL-2 concentration was measured by the Cytometric Bead Array (CBA) method. The results are shown in Table 3. When anti-CD80 and anti-CD86 antibodies were added, the IL-2 concentration was found to be significantly reduced from day 4 onwards during the 7-day culture period, compared to the negative control.
(MLR抑制評価)
上記7日間培養した細胞を25Gy放射線照射したドナー細胞(Batch#22TL290862:2.0x10^5 cells/well)とレシピエント細胞(Batch#23TL043676:2.0x10^5 cells/well)の共培養系に添加して96ウェルプレート(CORNING社)で培養した5日後の培養上清中のIFN-γとグランザイムBの濃度をCBA法で測定した。その結果を表4に示す。抗CD80と抗CD86抗体存在下に培養した細胞を添加することにより、細胞濃度依存的にIFN-γとグランザイムB濃度が低下していることから、MLRが抑制されたことが確認できた。 (MLR Suppression Evaluation)
The cells cultured for 7 days were added to a co-culture system of donor cells (Batch #22 TL290862: 2.0x10^5 cells/well) and recipient cells (Batch #23 TL043676: 2.0x10^5 cells/well) irradiated with 25 Gy, and the concentrations of IFN-γ and granzyme B in the culture supernatant 5 days after culturing in a 96-well plate (CORNING) were measured by the CBA method. The results are shown in Table 4. By adding cells cultured in the presence of anti-CD80 and anti-CD86 antibodies, the concentrations of IFN-γ and granzyme B were reduced in a cell concentration-dependent manner, confirming that MLR was suppressed.
上記7日間培養した細胞を25Gy放射線照射したドナー細胞(Batch#22TL290862:2.0x10^5 cells/well)とレシピエント細胞(Batch#23TL043676:2.0x10^5 cells/well)の共培養系に添加して96ウェルプレート(CORNING社)で培養した5日後の培養上清中のIFN-γとグランザイムBの濃度をCBA法で測定した。その結果を表4に示す。抗CD80と抗CD86抗体存在下に培養した細胞を添加することにより、細胞濃度依存的にIFN-γとグランザイムB濃度が低下していることから、MLRが抑制されたことが確認できた。 (MLR Suppression Evaluation)
The cells cultured for 7 days were added to a co-culture system of donor cells (Batch #22 TL290862: 2.0x10^5 cells/well) and recipient cells (Batch #23 TL043676: 2.0x10^5 cells/well) irradiated with 25 Gy, and the concentrations of IFN-γ and granzyme B in the culture supernatant 5 days after culturing in a 96-well plate (CORNING) were measured by the CBA method. The results are shown in Table 4. By adding cells cultured in the presence of anti-CD80 and anti-CD86 antibodies, the concentrations of IFN-γ and granzyme B were reduced in a cell concentration-dependent manner, confirming that MLR was suppressed.
(結果)
培養途中でIL-2を添加し至適濃度に調節した場合において、Tregのアロ特異的抑制能が増強した。 (result)
When IL-2 was added during the culture and adjusted to an optimal concentration, the allospecific suppressive ability of Treg was enhanced.
培養途中でIL-2を添加し至適濃度に調節した場合において、Tregのアロ特異的抑制能が増強した。 (result)
When IL-2 was added during the culture and adjusted to an optimal concentration, the allospecific suppressive ability of Treg was enhanced.
(注記)
以上のように、本開示の好ましい実施形態を用いて本開示を例示してきたが、本開示は、請求の範囲によってのみその範囲が解釈されるべきであることが理解される。本明細書において引用した特許、特許出願及び他の文献は、その内容自体が具体的に本明細書に記載されているのと同様にその内容が本明細書に対する参考として援用されるべきであることが理解される。本開示は、2023年1月30日に日本国特許庁に出願された特願2023―012172に対して優先権主張をするものであり、その内容は本出願においてすべての内容が本明細書を構成するものとして引用されることが理解される。 (Note)
As described above, the present disclosure has been illustrated using preferred embodiments of the present disclosure, but it is understood that the scope of the present disclosure should be interpreted only by the scope of the claims. It is understood that the patents, patent applications and other documents cited in this specification should be incorporated by reference to this specification in the same manner as if the contents themselves were specifically described in this specification. This disclosure claims priority to Japanese Patent Application No. 2023-012172 filed with the Japan Patent Office on January 30, 2023, and it is understood that the contents of the application are all cited as constituting the present specification.
以上のように、本開示の好ましい実施形態を用いて本開示を例示してきたが、本開示は、請求の範囲によってのみその範囲が解釈されるべきであることが理解される。本明細書において引用した特許、特許出願及び他の文献は、その内容自体が具体的に本明細書に記載されているのと同様にその内容が本明細書に対する参考として援用されるべきであることが理解される。本開示は、2023年1月30日に日本国特許庁に出願された特願2023―012172に対して優先権主張をするものであり、その内容は本出願においてすべての内容が本明細書を構成するものとして引用されることが理解される。 (Note)
As described above, the present disclosure has been illustrated using preferred embodiments of the present disclosure, but it is understood that the scope of the present disclosure should be interpreted only by the scope of the claims. It is understood that the patents, patent applications and other documents cited in this specification should be incorporated by reference to this specification in the same manner as if the contents themselves were specifically described in this specification. This disclosure claims priority to Japanese Patent Application No. 2023-012172 filed with the Japan Patent Office on January 30, 2023, and it is understood that the contents of the application are all cited as constituting the present specification.
製剤等に関連する産業(製薬)において利用可能な技術が提供される。
Technology that can be used in industries related to formulations (pharmaceuticals) will be provided.
Claims (14)
- 被験体由来の細胞から抗原特異的誘導型抑制性T細胞を選択的に誘導させる方法であって、抗原特異的誘導型抑制性T細胞を選択的に誘導する因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該被験体に由来しない抗原の含有物とを混合する工程を含む、方法。 A method for selectively inducing antigen-specific induced suppressor T cells from cells derived from a subject, the method comprising the step of mixing a factor that selectively induces antigen-specific induced suppressor T cells, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject or a substance containing an antigen not derived from the subject.
- 前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子である、請求項1に記載の方法。 The method according to claim 1, wherein the factor that selectively induces antigen-specific induced suppressor T cells is a regulatory factor that can regulate the production of IL-2 or a regulatory factor that can regulate the function of the produced IL-2.
- 前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、IL-2の産生を阻害することができる阻害因子または産生されたIL-2の機能を阻害することができる阻害因子である、請求項1に記載の方法。 The method according to claim 1, wherein the factor that selectively induces antigen-specific induced suppressor T cells is an inhibitory factor that can inhibit the production of IL-2 or an inhibitory factor that can inhibit the function of the produced IL-2.
- 前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、IL-2とIL-2受容体(IL-2R)との相互作用を阻害することができる阻害因子、ならびに/あるいは、CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子である、請求項1~3のいずれか一項に記載の方法。 The method according to any one of claims 1 to 3, wherein the factor that selectively induces antigen-specific induced suppressor T cells is an inhibitory factor capable of inhibiting the interaction between IL-2 and the IL-2 receptor (IL-2R) and/or an inhibitory factor capable of inhibiting the interaction between CD80 and/or CD86 and CD28.
- 前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、抗IL-2抗体、抗IL-2R抗体、抗IL-2Rα(CD25)抗体、抗IL-2Rβ(CD122)抗体またはこれらの抗原結合断片から選択される前記IL-2とIL-2Rとの相互作用を阻害することができる阻害因子である、請求項1~4のいずれか一項に記載の方法。 The method according to any one of claims 1 to 4, wherein the factor that selectively induces antigen-specific induced suppressor T cells is an inhibitor that can inhibit the interaction between IL-2 and IL-2R and is selected from anti-IL-2 antibodies, anti-IL-2R antibodies, anti-IL-2Rα (CD25) antibodies, anti-IL-2Rβ (CD122) antibodies, or antigen-binding fragments thereof.
- 前記抗原特異的誘導型抑制性T細胞を選択的に誘導する因子が、抗CD80抗体、抗CD86抗体、CD80およびCD86に対する二重特異性抗体、抗CD28抗体もしくはこれらの抗原結合断片、CTLA4-Ig融合タンパク質、およびCD28-Ig融合タンパク質からなる群より選択される前記CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子である、請求項1~5のいずれか一項に記載の方法。 The method according to any one of claims 1 to 5, wherein the factor that selectively induces antigen-specific induced suppressor T cells is an inhibitor that can inhibit the interaction between CD80 and/or CD86 and CD28, selected from the group consisting of anti-CD80 antibodies, anti-CD86 antibodies, bispecific antibodies against CD80 and CD86, anti-CD28 antibodies or antigen-binding fragments thereof, CTLA4-Ig fusion proteins, and CD28-Ig fusion proteins.
- 被験体由来の細胞から誘導型抑制性T細胞を作製する方法であって、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合する工程を含む、方法。 A method for producing induced suppressor T cells from cells derived from a subject, the method comprising the step of mixing a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, cells derived from the subject, and an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
- 前記IL-2の産生を調節することができる調節因子が、CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子、ならびにIL-2産生を阻害する小分子化合物から選択される、請求項7に記載の方法。 The method according to claim 7, wherein the regulator capable of regulating the production of IL-2 is selected from an inhibitor capable of inhibiting the interaction of CD80 and/or CD86 with CD28, and a small molecule compound that inhibits IL-2 production.
- 前記産生されたIL-2の機能を調節することができる調節因子が、IL-2とIL-2受容体(IL-2R)との相互作用を阻害することができる阻害因子、およびIL-2の作用を阻害する小分子化合物から選択される、請求項7に記載の方法。 The method according to claim 7, wherein the regulator capable of regulating the function of the produced IL-2 is selected from an inhibitor capable of inhibiting the interaction between IL-2 and the IL-2 receptor (IL-2R) and a small molecule compound that inhibits the action of IL-2.
- 前記CD80および/またはCD86と、CD28との相互作用を阻害することができる阻害因子は、抗CD80抗体、抗CD86抗体、CD80およびCD86に対する二重特異性抗体、抗CD28抗体もしくはこれらの抗原結合断片、CTLA4-Ig融合タンパク質、およびCD28-Ig融合タンパク質からなる群より選択される、請求項8に記載の方法。 The method according to claim 8, wherein the inhibitor capable of inhibiting the interaction between CD80 and/or CD86 and CD28 is selected from the group consisting of an anti-CD80 antibody, an anti-CD86 antibody, a bispecific antibody against CD80 and CD86, an anti-CD28 antibody or an antigen-binding fragment thereof, a CTLA4-Ig fusion protein, and a CD28-Ig fusion protein.
- 前記IL-2とIL-2受容体(IL-2R)との相互作用を阻害することができる阻害因子が、抗IL-2抗体、抗IL-2R抗体、抗IL-2Rα(CD25)抗体、抗IL-2Rβ(CD122)抗体またはこれらの抗原結合断片から選択される、請求項9に記載の方法。 The method according to claim 9, wherein the inhibitor capable of inhibiting the interaction between IL-2 and IL-2 receptor (IL-2R) is selected from an anti-IL-2 antibody, an anti-IL-2R antibody, an anti-IL-2Rα (CD25) antibody, an anti-IL-2Rβ (CD122) antibody, or an antigen-binding fragment thereof.
- 被験体由来の細胞から抗原特異的誘導型抑制性T細胞を選択的に誘導させるための組成物であって、該組成物は、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子を含み、該組成物が、該被験体由来の細胞、および該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物と接触させられることを特徴とする、組成物。 A composition for selectively inducing antigen-specific inducible suppressor T cells from cells derived from a subject, the composition comprising a regulator capable of regulating the production of IL-2 or a regulator capable of regulating the function of the produced IL-2, the composition being contacted with cells derived from the subject and an antigen derived from the subject or an antigen not derived from the subject or a substance containing the antigen.
- 免疫応答により媒介される被験体の疾患を処置するための組成物であって、該組成物が、IL-2の産生を調節することができる調節因子または産生されたIL-2の機能を調節することができる調節因子と、該被験体由来の細胞と、該被験体由来の抗原あるいは該被験体に由来しない抗原または該抗原の含有物とを混合することによって選択的に誘導された抗原特異的誘導型抑制性T細胞を含む、組成物。 A composition for treating a disease in a subject mediated by an immune response, the composition comprising a regulatory factor capable of regulating the production of IL-2 or a regulatory factor capable of regulating the function of the produced IL-2, and antigen-specific induced suppressor T cells selectively induced by mixing cells derived from the subject with an antigen derived from the subject or an antigen not derived from the subject, or a substance containing the antigen.
- 前記疾患が、移植免疫拒絶反応、アレルギー、自己免疫疾患、移植片対宿主病、およびiPS細胞もしくはES細胞またはそれらから分化した細胞、組織もしくは臓器の移植によって引き起こされる免疫拒絶反応からなる群より選択される、請求項13に記載の組成物。 The composition according to claim 13, wherein the disease is selected from the group consisting of transplant immune rejection, allergy, autoimmune disease, graft-versus-host disease, and immune rejection caused by transplantation of iPS cells or ES cells, or cells, tissues, or organs differentiated therefrom.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2023012172 | 2023-01-30 | ||
JP2023-012172 | 2023-01-30 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024162262A1 true WO2024162262A1 (en) | 2024-08-08 |
Family
ID=92146788
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2024/002648 WO2024162262A1 (en) | 2023-01-30 | 2024-01-29 | Method for selectively inducing antigen-specific inductive regulatory t cells |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024162262A1 (en) |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2016520081A (en) * | 2013-05-17 | 2016-07-11 | エービーウィズ バイオ,インク. | Regulatory T cells and uses thereof |
-
2024
- 2024-01-29 WO PCT/JP2024/002648 patent/WO2024162262A1/en unknown
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2016520081A (en) * | 2013-05-17 | 2016-07-11 | エービーウィズ バイオ,インク. | Regulatory T cells and uses thereof |
Non-Patent Citations (1)
Title |
---|
JAMIE B. SPANGLER, JAKUB TOMALA, VINCENT C. LUCA, KEVIN M. JUDE, SHEN DONG, AARON M. RING, PETRA VOTAVOVA, MARION PEPPER, MAREK KO: "Antibodies to Interleukin-2 Elicit Selective T Cell Subset Potentiation through Distinct Conformational Mechanisms", IMMUNITY, CELL PRESS, AMSTERDAM, NL, vol. 42, no. 5, 1 May 2015 (2015-05-01), AMSTERDAM, NL , pages 815 - 825, XP055336250, ISSN: 1074-7613, DOI: 10.1016/j.immuni.2015.04.015 * |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7451627B2 (en) | Chimeric receptor and its use | |
CN111479613A (en) | Methods of administering chimeric antigen receptor immunotherapy | |
CN118662516A (en) | Methods of administering chimeric antigen receptor immunotherapy | |
WO2022166947A1 (en) | Preparation method for tumor-infiltrating lymphocytes and use thereof | |
JP2022513062A (en) | Methods of Dosing Manipulated T Cells to Treat B-Cell Malignancies | |
JP2022028075A (en) | Antibodies that induce immune tolerance, induced lymphocytes, and cell therapeutic agent and therapeutic method using induced lymphocytes | |
JP2024015377A (en) | Antibody capable of inducing immune tolerance produced using cell mixture having complexed state, and induced lymphocyte or cell therapeutic agent and cell therapy method each using induced lymphocyte | |
WO2024162262A1 (en) | Method for selectively inducing antigen-specific inductive regulatory t cells | |
TWI832868B (en) | Compositions for inducing infectious immune tolerance | |
WO2023190942A1 (en) | Method for evaluating quality of inducement inhibitory-t-cell formulation | |
RU2816592C2 (en) | Immune tolerance inducing antibodies, induced lymphocytes and therapeutic agent/method of cell therapy using induced lymphocytes | |
TWI854983B (en) | Antibodies that induce immune tolerance, induced lymphocytes, and cell therapy methods using induced lymphocytes | |
US12139537B2 (en) | Antibody inducing immune tolerance, induced lymphocyte, and cell therapy agent therapeutic method using induced lymphocyte | |
JP2024160293A (en) | Antibodies that induce immune tolerance, induced lymphocytes, and cell therapy using induced lymphocytes | |
TWI837437B (en) | Chimeric antigen receptor t cell therapy | |
TWI855499B (en) | Chimeric receptors and methods of use thereof | |
Kostopoulos | Radiation-Driven cross presentation and STING activation enhance CAR T cell therapy in a murine lymphoma model | |
JP2023526228A (en) | Methods for treating glioblastoma | |
JP2024504397A (en) | Formulations and methods for CAR T cell medicines |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24750211 Country of ref document: EP Kind code of ref document: A1 |